WO2014204578A1 - Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing - Google Patents

Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing Download PDF

Info

Publication number
WO2014204578A1
WO2014204578A1 PCT/US2014/035162 US2014035162W WO2014204578A1 WO 2014204578 A1 WO2014204578 A1 WO 2014204578A1 US 2014035162 W US2014035162 W US 2014035162W WO 2014204578 A1 WO2014204578 A1 WO 2014204578A1
Authority
WO
WIPO (PCT)
Prior art keywords
target
rna
cas9
grna
sites
Prior art date
Application number
PCT/US2014/035162
Other languages
French (fr)
Inventor
J. Keith Joung
Shengdar TSAI
Original Assignee
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2014/028630 external-priority patent/WO2014144288A1/en
Application filed by The General Hospital Corporation filed Critical The General Hospital Corporation
Priority to US14/900,444 priority Critical patent/US10011850B2/en
Priority to US15/107,550 priority patent/US10526589B2/en
Priority to KR1020167020111A priority patent/KR20160102056A/en
Priority to CA2935032A priority patent/CA2935032C/en
Priority to EP21191144.1A priority patent/EP3985124A1/en
Priority to AU2014370416A priority patent/AU2014370416B2/en
Priority to EP14875819.6A priority patent/EP3090044B1/en
Priority to PCT/US2014/056416 priority patent/WO2015099850A1/en
Priority to CN201480076396.6A priority patent/CN106103706B/en
Priority to JP2016542968A priority patent/JP6721508B2/en
Priority to CN202110920229.7A priority patent/CN113684205A/en
Publication of WO2014204578A1 publication Critical patent/WO2014204578A1/en
Priority to US16/003,973 priority patent/US10544433B2/en
Priority to JP2019218086A priority patent/JP7005580B2/en
Priority to US16/735,146 priority patent/US20200165587A1/en
Priority to US16/751,578 priority patent/US11098326B2/en
Priority to AU2021203309A priority patent/AU2021203309B2/en
Priority to AU2023258349A priority patent/AU2023258349A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/21Endodeoxyribonucleases producing 5'-phosphomonoesters (3.1.21)
    • C12Y301/21004Type II site-specific deoxyribonuclease (3.1.21.4)

Definitions

  • RNA-guided Fokl Nucleases RNNs
  • RNA-guided genome editing e.g., editing using CRISPR/Cas9 systems
  • FNNs RNA-guided Fokl Nucleases
  • FokI-dCas9 fusion proteins e.g., FokI-dCas9 fusion proteins
  • CRISPR clustered, regularly interspaced, short palindromic repeats
  • Cas CRISPR-associated systems
  • the Cas9 nuclease from S. pyogenes can be guided via base pair complementarity between the first 20 nucleotides of an engineered gRNA and the complementary strand of a target genomic DNA sequence of interest that lies next to a protospacer adjacent motif (PAM), e.g., a PAM matching the sequence NGG or NAG (Shen et al, Cell Res (2013); Dicarlo et al, Nucleic Acids Res (2013); Jiang et al, Nat Biotechnol 31 , 233-239 (2013); Jinek et al, Elife 2, e00471 (2013); Hwang et al, Nat Biotechnol 31, 227-229 (2013); Cong et al, Science 339, 819-823 (2013); Mali et al, Science 339, 823-826 (2013c); Cho et al, Nat
  • CRISPR-Cas nucleases can tolerate up to five mismatches and still cleave; it is hard to predict the effects of any given single or combination of mismatches on activity. Taken together, these nucleases can show significant off-target effects but it can be challenging to predict these sites. Described herein are methods for increasing the specificity of genome editing using the
  • RNA-guided Fokl Nucleases e.g., Fokl- Cas9 or FokI-dCas9-based fusion proteins.
  • the invention provides FokI-dCas9 fusion proteins, comprising a Fokl catalytic domain sequence fused to the terminus, e.g., the N terminus, of dCas9, optionally with an intervening linker, e.g., a linker of from 2-30 amino acids, e.g., 4-12 amino acids, e.g., Gly 4 Ser.
  • the Fokl catalytic domain comprises amino acids 388-583 or 408-583 of SEQ ID NO:4.
  • the dCas9 comprises mutations at the dCas9 comprises mutations at D10, E762, H983, or D986; and at H840 or N863; e.g., at: (i) DIOA or DION; and (ii) H840A, H840Y or H840N.
  • the invention provides nucleic acids encoding these fusion proteins, vector comprising the nucleic acids, and host cells harboring or expressing the nucleic acids, vectors, or fusion proteins.
  • the invention provides methods for inducing a sequence- specific break in a double-stranded DNA molecule, e.g., in a genomic sequence in a cell, the method comprising expressing in the cell, or contacting the cell with, the FokI-dCas9 fusion protein described herein, and:
  • each of the two single guide R As include sequences that are each complementary to one strand of the target sequence such that using both guide RNAs results in targeting both strands (i.e., one single guide RNA targets a first strand, and the other guide RNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double-stranded break, or
  • each of the two crRNAs include sequences that are complementary to one strand of the target sequence such that using both crRNAs results in targeting both strands (i.e., one crRNA targets a first strand, and the other crRNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double- stranded break.
  • the invention provides methods for increasing specificity of RNA-guided genome editing in a cell, the method comprising contacting the cell with an RNA-guided Fokl Nuclease (RFN) fusion protein described herein.
  • RFN RNA-guided Fokl Nuclease
  • the method may further comprise expressing in the cell, or contacting the cell with, (a) two single guide RNAs, wherein each of the two single guide RNAs include sequences that are each complementary to one strand of the target sequence such that using both guide RNAs results in targeting both strands (i.e., one single guide RNA targets a first strand, and the other guide RNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double-stranded break, or
  • each of the two crRNAs include sequences that are complementary to one strand of the target sequence such that using both crRNAs results in targeting both strands (i.e., one crRNA targets a first strand, and the other crRNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double- stranded break.
  • the two target genomic sequences i.e., the sequences to which the target complementarity regions of the crR A or single guide R As are complementary
  • an indel mutation is induced between the two target sequences.
  • the specificity of RNA-guided genome editing in a cell is increased.
  • Figure 1 Schematic illustrating a gR A/Cas9 nuclease complex bound to its target DNA site. Scissors indicate approximate cleavage points of the Cas9 nuclease on the genomic DNA target site. Note the numbering of nucleotides on the guide RNA proceeds in an inverse fashion from 5' to 3'.
  • FIG. 2B Schematic overview of the EGFP disruption assay. Repair of targeted Cas9-mediated double-stranded breaks in a single integrated EGFP-PEST reporter gene by error-prone NHEJ-mediated repair leads to frame-shift mutations that disrupt the coding sequence and associated loss of fluorescence in cells.
  • Figures 2C-F Activities of RGNs harboring sgRNAs bearing (C) single mismatches, (D) adjacent double mismatches, (E) variably spaced double mismatches, and (F) increasing numbers of adjacent mismatches assayed on three different target sites in the EGFP reporter gene sequence. Mean activities of replicates (see Online Methods) are shown, normalized to the activity of a perfectly matched gRNA. Error bars indicate standard errors of the mean. Positions mismatched in each gRNA are highlighted in grey in the grid below. Sequences of the three EGFP target sites were as follows:
  • EGFP Site 1 GGGCACGGGCAGCTTGCCGGTGG (SEQ ID NO : 1 )
  • EGFP Site 2 GATGCCGTTCTTCTGCTTGTCGG (SEQ ID NO:2)
  • EGFP Site 3 GGTGGTGCAGATGAACTTCAGGG (SEQ ID NO:3)
  • Figure 2G Mismatches at the 5' end of the gRNA make CRISPR/Cas more sensitive more 3' mismatches.
  • the gRNAs Watson-Crick base pair between the RNA&DNA with the exception of positions indicated with an "m" which are mismatched using the Watson-Crick transversion (i.e. EGFP Site#2 M18-19 is mismatched by changing the gRNA to its Watson-Crick partner at positions 18 & 19.
  • positions near the 5 ' of the gRNA are generally very well tolerated, matches in these positions are important for nuclease activity when other residues are mismatched.
  • Figure 2H Efficiency of Cas9 nuclease activities directed by gRNAs bearing variable length complementarity regions ranging from 15 to 25 nts in a human cell- based U20S EGFP disruption assay. Expression of a gRNA from the U6 promoter requires the presence of a 5 ' G and therefore it was only possible to evaluate gRNAs harboring certain lengths of complementarity to the target DNA site (15, 17, 19, 20, 21, 23, and 25 nts).
  • Figure 3B Efficiencies of targeted indel mutations introduced at seven different human endogenous gene targets by matched standard and tru-RGNs.
  • Figure 3C DNA sequences of indel mutations induced by RGNs using a tru- gRNA or a matched full-length gRNA targeted to the EMXl site.
  • the portion of the target DNA site that interacts with the gRNA complementarity region is highlighted in grey with the first base of the PAM sequence shown in lowercase. Deletions are indicated by dashes highlighted in grey and insertions by italicized letters highlighted in grey. The net number of bases deleted or inserted and the number of times each sequence was isolated are shown to the right.
  • FIG. 3E U20S.EGFP cells were transfected with variable amounts of full- length gRNA expression plasmids (top) or tru-gRNA expression plasmids (bottom) together with a fixed amount of Cas9 expression plasmid and then assayed for percentage of cells with decreased EGFP expression. Mean values from duplicate experiments are shown with standard errors of the mean. Note that the data obtained with tru-gRNA matches closely with data from experiments performed with full- length gRNA expression plasmids instead of tru-gRNA plasmids for these three EGFP target sites.
  • Figure 3F U20S.EGFP cells were transfected with variable amount of Cas9 expression plasmid together with variable amounts of full-length gRNA expression plasmids (top) or tru-gRNA expression plasmids (bottom) (amounts determined for each tru-gRNA from the experiments of Figure 3E). Mean values from duplicate experiments are shown with standard errors of the mean. Note that the data obtained with tru-gRNA matches closely with data from experiments performed with full- length gRNA expression plasmids instead of tru-gRNA plasmids for these three EGFP target sites. The results of these titrations determined the concentrations of plasmids used in the EGFP disruption assays performed in Examples 1 and 2.
  • Figures 4A-C RNA-guided Fokl nucleases and a CRISPR/Cas Subtype Ypest protein 4 (Csy4)-based multiplex gRNA expression system.
  • FIG. 1 Schematic overview of a Csy4-based multiplex gRNA expression system.
  • Two gRNAs (with any 5 ' end nucleotide) are co-expressed in a single transcript from a U6 promoter with each gRNA flanked by Csy4 recognition sites.
  • Csy4 cleaves and releases gRNAs from the transcript.
  • the Csy4 recognition site remains at the 3 ' end of the gRNA with a Csy4 nuclease bound to that site.
  • Figures 5A-F Design and optimization of RNA-guided Fokl nucleases.
  • FIGS 6A-D Dimerization of FokI-dCas9 RFNs is required for efficient genome editing activity.
  • MLHl mutL homo log 1
  • FIGS 7A-B Mutagenic activities of a Cas9 nickase or FokI-dCas9 co- expressed with a single gRNA.
  • Indel mutation frequencies were determined by deep sequencing. Each indel frequency value reported was determined from a single deep sequencing library prepared from genomic DNA pooled from three independent transfection
  • VEGFA Vascular Endothelial Growth Factor A
  • DDB2 Damage- Specific DNA Binding Protein 2
  • FANCF Fanconi Anemia, Complementation Group F
  • FES Feline Sarcoma Oncogene
  • RUNX Runt-Related Transcription Factor 1.
  • Figures 8A-C Single Cas9 nickases can introduce point mutations with high efficiencies into their target sites.
  • RGNs CRISPR RNA-guided nucleases
  • T7EI assay (which, as performed in our laboratory, has a reliable detection limit of ⁇ 2 to 5% mutation frequency). Because these mutation rates were very high, it was possible to avoid using deep sequencing methods previously required to detect much lower frequency ZFN- and TALEN-induced off-target alterations (Pattanayak et al., Nat Methods 8, 765-770 (2011); Perez et al, Nat Biotechnol 26, 808-816 (2008);
  • off-target sites were seen for a number of RGNs, identification of these sites was neither comprehensive nor genome -wide in scale. For the six RGNs studied, only a very small subset of the much larger total number of potential off-target sequences in the human genome was examined. Although examining such large numbers of loci for off-target mutations by T7EI assay is neither a practical nor a cost-effective strategy, the use of high-throughput sequencing in future studies might enable the interrogation of larger numbers of candidate off-target sites and provide a more sensitive method for detecting bona fide off-target mutations. For example, such an approach might enable the unveiling of additional off-target sites for the two RGNs for which we failed to uncover any off-target mutations.
  • a number of strategies can be used to minimize the frequencies of genomic off-target mutations.
  • the specific choice of RGN target site can be optimized; given that off-target sites that differ at up to five positions from the intended target site can be efficiently mutated by RGNs, choosing target sites with minimal numbers of off-target sites as judged by mismatch counting seems unlikely to be effective; thousands of potential off-target sites that differ by four or five positions within the 20 bp RNA:DNA complementarity region will typically exist for any given RGN targeted to a sequence in the human genome. It is also possible that the nucleotide content of the gRNA complementarity region might influence the range of potential off-target effects.
  • RNA:DNA hybrids For example, high GC-content has been shown to stabilize RNA:DNA hybrids (Sugimoto et al, Biochemistry 34, 1121 1-11216 (1995)) and therefore might also be expected to make gRNA/genomic DNA hybridization more stable and more tolerant to mismatches. Additional experiments with larger numbers of gRNAs will be needed to assess if and how these two parameters (numbers of mismatched sites in the genome and stability of the RNA:DNA hybrid) influence the genome-wide specificities of RGNs. However, it is important to note that even if such predictive parameters can be defined, the effect of implementing such guidelines would be to further restrict the targeting range of RGNs.
  • RGN-induced off-target effects might be to reduce the concentrations of gRNA and Cas9 nuclease expressed in the cell. This idea was tested using the RGNs for VEGFA target sites 2 and 3 in
  • CRISPR-Cas RNA-guided nucleases based on the S.
  • pyogenes Cas9 protein can have significant off-target mutagenic effects that are comparable to or higher than the intended on-target activity (Example 1). Such off-target effects can be problematic for research and in particular for potential therapeutic applications. Therefore, methods for improving the specificity of
  • RGNs RNA guided nucleases
  • Cas9 RGNs can induce high-frequency indel mutations at off-target sites in human cells (see also Cradick et al., 2013; Fu et al., 2013; Hsu et al., 2013; Pattanayak et al, 2013). These undesired alterations can occur at genomic sequences that differ by as many as five mismatches from the intended on- target site (see Example 1).
  • RNA-guided nucleases are to be used for research and therapeutic applications.
  • Dimerization is an attractive potential strategy for improving the specificity of Cas9 nucleases. This is distinct from a paired Cas9 nickase approach, which is not a true dimeric system. Paired nickases work by co-localizing two Cas9 nickases on a segment of DNA, thereby inducing high efficiency genome editing via an undefined mechanism. Because dimerization is not required for enzymatic activity, single Cas9 nickases can also induce indels with high efficiencies at certain sites (via an unknown mechanism) and can therefore potentially cause unwanted off-target mutations in the genome.
  • one strategy to improve the specificity of RGNs is fusing a Fokl endonuclease domain to a catalytically inactive form of Cas9 bearing the D10A and H840A mutations (also known as dCas9).
  • Fokl nuclease domain functions as a dimer and therefore two subunits must be recruited to the same local piece of DNA in order to induce a double-stranded break.
  • Figure 9A and Example 2 two FokI-dCas9 fusions are recruited in an appropriate configuration using two different gRNAs to yield a double-stranded break.
  • the Fokl- dCas9 fusions would bind to a site that is twice as long as that of a single RGN and therefore this system would be expected to be more specific.
  • FokI-dCas9 fusion proteins wherein the Fokl sequence is fused to dCas9 (preferably to the amino-terminal end of dCas9, but also optionally to the carboxy terminus), optionally with an intervening linker, e.g., a linker of from 2-30 amino acids, e.g., 4-12 amino acids, e.g., Gly 4 Ser (SEQ ID NO:23) or (Gly 4 Ser) 3 .
  • the fusion proteins include a linker between the dCas9 and the Fokl domains.
  • Linkers that can be used in these fusion proteins (or between fusion proteins in a concatenated structure) can include any sequence that does not interfere with the function of the fusion proteins.
  • the linkers are short, e.g., 2-20 amino acids, and are typically flexible (i.e., comprising amino acids with a high degree of freedom such as glycine, alanine, and serine).
  • the linker comprises one or more units consisting of GGGS (SEQ ID NO:22) or GGGGS (SEQ ID NO:23), e.g., two, three, four, or more repeats of the GGGS (SEQ ID NO:22) or GGGGS (SEQ ID NO:23) unit.
  • linker sequences can also be used.
  • a RNA-guided Fokl nuclease platform in which dimerization, rather than just co-localization, is required for efficient genome editing activity. These nucleases can robustly mediate highly efficient genome editing in human cells and can reduce off-target mutations to undetectable levels as judged by sensitive deep sequencing methods. Also described is an efficient system for expressing pairs of gRNAs with any 5 ' end nucleotide, a method that confers a wider targeting range on the RFN platform. Finally, monomeric Cas9 nickases generally introduce more undesirable indels and point mutations than the nucleases described herein in the presence of a single gRNA.
  • RNA-guided Fokl Nuclease (RFN) platform for performing robust and highly specific genome editing in human cells.
  • RFNs require two gRNAs for activity and function as dimers.
  • the engineering of an active RFN required fusion of the Fokl nuclease domain to the amino-terminal end of the dCas9 protein, an architecture different from ZFNs and TALENs in which the Fokl domain is fused to the carboxy-terminal end of engineered zinc finger or transcription activator-like effector repeat arrays.
  • RFNs also require that the half-sites bound by each Fok-dCas9/gRNA complex have a particular relative orientation (PAMs out) with a relatively restricted intervening spacer length of 14 to 17 bps (although activity may be possible at additional spacings but with less consistent success).
  • PAMs out relative orientation
  • RFNs The dimeric nature of RFNs provides important specificity advantages relative to standard monomeric Cas9 nucleases. In an ideal dimeric system, little to no activity will be observed with monomers on half-sites.
  • the present data demonstrate that FokI-dCas9 directed by a single gRNA induces very little or no mutagenesis at RFN half-sites. 12 single gRNAs (for six RFN target sites) were tested with co- expressed FokI-dCas9 and indels were observed at very low frequencies (range of 0.0045% to 0.47%), in some cases at levels as low as background rates observed in control cells in which there was no expression of gRNA or nuclease.
  • Fokl nuclease domain functions as a dimer
  • any indels observed with a single gR A are likely due to recruitment of a FokI-dCas9 dimer to the DNA.
  • FokI-dCas9 dimer Regardless of mechanism, given that only very low level mutagenesis was observed when FokI-dCas9 was tested with single gRNAs at 12 on-target half-sites, it is very unlikely that any mutagenesis will be induced at partially mismatched, off-target half- sites. Indeed, an RFN targeted to VEGFA did not induce detectable mutations at known off-target sites of one of the gRNAs as judged by deep sequencing.
  • RFNs are a true dimeric system, they possess a number of important advantages over paired nickase technology, which depends on co-localization but does not require dimerization.
  • paired Cas9 nickases show greater promiscuity in the orientation and spacing of target half-sites than dimeric RFNs and therefore have a greater potential range of sites at which off-target mutations might be induced.
  • Paired nickase half-sites can be oriented with their PAMs in or PAMs out and with spacer sequences ranging in length from 0 to 1000 bps (Ran et al, Cell 154, 1380-1389 (2013); Mali et al, Nat Biotechnol 31, 833-838 (2013); Cho et al, Genome Res (2013)).
  • This promiscuity exists because the genome editing activities of Cas9 nickases do not depend on dimerization of the enzyme but rather just co- localization of the two nicks.
  • RFNs are much more stringent in their specificities— half-sites must have their PAMs out and must be spaced apart by 14 to 17 bps, due to the requirement for two appropriately positioned Fokl cleavage domains for efficient cleavage.
  • Fokl is a type lis restriction endonuclease that includes a DNA recognition domain and a catalytic (endonuclease) domain.
  • the fusion proteins described herein can include all of Fokl or just the catalytic endonuclease domain, e.g., amino acids 388-583 or 408-583 of GenBank Acc. No. AAA24927.1, e.g., as described in Li et al, Nucleic Acids Res. 39(1): 359-372 (2011); Cathomen and Joung, Mol. Ther. 16: 1200-1207 (2008), or a mutated form of Fokl as described in Miller et al. Nat
  • An exemplary nucleic acid sequence encoding Fokl is as follows:
  • the Fokl nuclease used herein is at least about 50% identical SEQ ID NO:4, e.g., to amino acids 388-583 or 408-583 of SEQ ID NO:4. These variant nucleases must retain the ability to cleave DNA.
  • the nucleotide sequences are about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identical to amino acids 388-583 or 408-583 of SEQ ID NO:4. In some embodiments, any differences from amino acids 388-583 or 408-583 of SEQ ID NO:4 are in non-conserved regions.
  • the sequences are aligned for optimal comparison purposes (gaps are introduced in one or both of a first and a second amino acid or nucleic acid sequence as required for optimal alignment, and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 50%> (in some embodiments, about 50%, 55%, 60%, 65%, 70%, 75%, 85%, 90%, 95%, or 100% of the length of the reference sequence is aligned).
  • the nucleotides or residues at corresponding positions are then compared. When a position in the first sequence is occupied by the same nucleotide or residue as the corresponding position in the second sequence, then the molecules are identical at that position.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package, using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.
  • a number of bacteria express Cas9 protein variants.
  • Streptococcus pyogenes is presently the most commonly used; some of the other Cas9 proteins have high levels of sequence identity with the S. pyogenes Cas9 and use the same guide R As. Others are more diverse, use different gR As, and recognize different PAM sequences as well (the 2-5 nucleotide sequence specified by the protein which is adjacent to the sequence specified by the RNA). Chylinski et al. classified Cas9 proteins from a large group of bacteria (RNA Biology 10:5, 1-12; 2013), and a large number of Cas9 proteins are listed in supplementary figure 1 and supplementary table 1 thereof, which are incorporated by reference herein. Additional Cas9 proteins are described in Esvelt et al., Nat Methods.
  • Cas9 molecules of a variety of species can be used in the methods and compositions described herein. While the S. pyogenes and S. thermophilus Cas9 molecules are the subject of much of the disclosure herein, Cas9 molecules of, derived from, or based on the Cas9 proteins of other species listed herein can be used as well. In other words, while the much of the description herein uses S. pyogenes and S. thermophilus Cas9 molecules, Cas9 molecules from the other species can replace them. Such species include those set forth in the following table, which was created based on supplementary figure 1 of Chylinski et al, 2013.
  • the constructs and methods described herein can include the use of any of those Cas9 proteins, and their corresponding guide RNAs or other guide RNAs that are compatible.
  • the Cas9 from Streptococcus thermophilus LMD-9 CRISPR1 system has also been shown to function in human cells in Cong et al (Science 339, 819 (2013)).
  • Cas9 orthologs from N. meningitides are described in Hou et al, Proc Natl Acad Sci U S A. 2013 Sep 24;110(39): 15644-9 and Esvelt et al, Nat Methods. 2013 Nov;10(l 1): 1116-21. Additionally, Jinek et al. showed in vitro that Cas9 orthologs from S. thermophilus and L.
  • innocua (but not from N. meningitidis or C. jejuni, which likely use a different guide RNA), can be guided by a dual S. pyogenes gRNA to cleave target plasmid DNA, albeit with slightly decreased efficiency.
  • the present system utilizes the Cas9 protein from S. pyogenes, either as encoded in bacteria or codon-optimized for expression in mammalian cells, containing mutations at D10, E762, H983, or D986 and H840 or N863, e.g., D10A/D10N and H840A/H840N/H840Y, to render the nuclease portion of the protein catalytically inactive; substitutions at these positions could be alanine (as they are in Nishimasu al, Cell 156, 935-949 (2014)) or they could be other residues, e.g., glutamine, asparagine, tyrosine, serine, or aspartate, e.g.,, E762Q, H983N, H983Y, D986N, N863D, N863S, or N863H ( Figure 1C).
  • S. pyogenes either as encoded in bacteria or codon-optimized for expression in
  • sequence of the catalytically inactive S. pyogenes Cas9 that can be used in the methods and compositions described herein is as follows; the exemplary mutations of D10A and H840A are in bold and underlined. 10 20 30 40 50 60
  • PAAFKYFDTT IDRKRYTSTK EVLDATLIHQ SITGLYETRI DLSQLGGD SEQ ID NO: 5
  • the Cas9 nuclease used herein is at least about 50% identical to the sequence of S. pyogenes Cas9, i.e., at least 50% identical to SEQ ID NO:5.
  • the nucleotide sequences are about 50%>, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identical to SEQ ID NO:5.
  • any differences from SEQ ID NO:5 are in non-conserved regions, as identified by sequence alignment of sequences set forth in Chylinski et al, RNA Biology 10:5, 1-12; 2013 (e.g., in supplementary figure 1 and supplementary table 1 thereof); Esvelt et al, Nat Methods. 2013 Nov; 10(11): 1116-21 and Fonfara et al, Nucl. Acids Res. (2014) 42 (4): 2577-2590. [Epub ahead of print 2013 Nov 22] doi: 10.1093/nar/gktl074. Identity is determined as set forth above.
  • gRNAs Guide RNAs
  • RNAs generally speaking come in two different systems: System 1, which uses separate crRNA and tracrRNAs that function together to guide cleavage by Cas9, and System 2, which uses a chimeric crRNA-tracrRNA hybrid that combines the two separate guide RNAs in a single system (referred to as a single guide RNA or sgRNA, see also Jinek et al., Science 2012; 337:816-821).
  • the tracrRNA can be variably truncated and a range of lengths has been shown to function in both the separate system (system 1) and the chimeric gRNA system (system 2).
  • tracrRNA may be truncated from its 3' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts.
  • the tracrRNA molecule may be truncated from its 5' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts.
  • the tracrRNA molecule may be truncated from both the 5' and 3' end, e.g., by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 nts on the 5' end and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts on the 3' end.
  • vectors e.g., plasmids
  • plasmids encoding more than one gRNA are used, e.g., plasmids encoding, 2, 3, 4, 5, or more gRNAs directed to different sites in the same region of the target gene.
  • Cas9 nuclease can be guided to specific 17-20 nt genomic targets bearing an additional proximal protospacer adjacent motif (PAM), e.g., of sequence NGG, using a guide RNA, e.g., a single gRNA or a tracrRNA/crRNA, bearing 17-20 nts at its 5' end that are complementary to the complementary strand of the genomic DNA target site.
  • PAM proximal protospacer adjacent motif
  • the present methods can include the use of a single guide RNA comprising a crRNA fused to a normally trans-encoded tracrRNA, e.g., a single Cas9 guide RNA as described in Mali et al, Science 2013 Feb 15; 339(6121):823-6, with a sequence at the 5' end that is complementary to the target sequence, e.g., of 25-17, optionally 20 or fewer nucleotides (nts), e.g., 20, 19, 18, or 17 nts, preferably 17 or 18 nts, of the complementary strand to a target sequence immediately 5 Of a protospacer adjacent motif (PAM), e.g., NGG, NAG, or NNGG
  • the single Cas9 guide RNA consists of the sequence:
  • X17-20 is the nucleotide sequence complementary to 17-20 consecutive nucleotides of the target sequence.
  • DNAs encoding the single guide RNAs have been described previously in the literature (Jinek et al, Science. 337(6096):816-21 (2012) and Jinek et al, Elife. 2:e00471 (2013)).
  • the guide RNAs can include X N which can be any sequence, wherein N (in the RNA) can be 0-200, e.g., 0-100, 0-50, or 0-20, that does not interfere with the binding of the ribonucleic acid to Cas9.
  • the guide RNA includes one or more Adenine (A) or Uracil (U) nucleotides on the 3 ' end.
  • the RNA includes one or more U, e.g., 1 to 8 or more Us (e.g., U, UU, UUU, UUUU, UUUUU, UUUUU, UUUUUU, UUUUUU, UUUUUU, UUUUUUUUUU, UUUUUUUUUUU) at the 3' end of the molecule, as a result of the optional presence of one or more Ts used as a termination signal to terminate RNA PolIII transcription.
  • gRNA e.g., the crRNA and tracrRNA found in naturally occurring systems.
  • a single tracrRNA would be used in conjunction with multiple different crRNAs expressed using the present system, e.g., the following:
  • the methods include contacting the cell with a tracrRNA comprising or consisting of the sequence
  • the tracrRNA molecule may be truncated from its 3' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts.
  • the tracrR A molecule may be truncated from its 5 ' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts.
  • the tracrRNA molecule may be truncated from both the 5' and 3' end, e.g., by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 nts on the 5' end and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts on the 3' end.
  • Exemplary tracrRNA sequences in addition to SEQ ID NO: 8 include the following:
  • GGCACCGAGUCGGUGC SEQ ID NO: 18 or an active portion thereof.
  • tracrRNA (SEQ ID NO: 14) is used as a crRNA, the following tracrRNA is used:
  • tracrRNA is used as a crRNA, the following tracrRNA is used:
  • the gRNA is targeted to a site that is at least three or more mismatches different from any sequence in the rest of the genome in order to minimize off-target effects.
  • RNA oligonucleotides such as locked nucleic acids (LNAs) have been demonstrated to increase the specificity of RNA-DNA hybridization by locking the modified oligonucleotides in a more favorable (stable) conformation.
  • LNAs locked nucleic acids
  • 2'-0-methyl RNA is a modified base where there is an additional covalent linkage between the 2' oxygen and 4' carbon which when incorporated into oligonucleotides can improve overall thermal stability and selectivity (Formula I).
  • the tru-gRNAs disclosed herein may comprise one or more modified RNA oligonucleotides.
  • the truncated guide RNAs molecules described herein can have one, some or all of the 17-18 or 17-19 nts 5 ' region of the guideRNA complementary to the target sequence are modified, e.g., locked (2'-0-4'-C methylene bridge), 5'-methylcytidine, 2'-0-methyl-pseudouridine, or in which the ribose phosphate backbone has been replaced by a polyamide chain (peptide nucleic acid), e.g., a synthetic ribonucleic acid.
  • a polyamide chain peptide nucleic acid
  • one, some or all of the nucleotides of the tru-gRNA sequence may be modified, e.g., locked (2'-0-4'-C methylene bridge), 5'- methylcytidine, 2'-0-methyl-pseudouridine, or in which the ribose phosphate backbone has been replaced by a polyamide chain (peptide nucleic acid), e.g., a synthetic ribonucleic acid.
  • the single guide RNAs and/or crRNAs and/or tracrRNAs can include one or more Adenine (A) or Uracil (U) nucleotides on the 3 ' end.
  • A Adenine
  • U Uracil
  • RNA-DNA heteroduplexes can form a more promiscuous range of structures than their DNA-DNA counterparts.
  • DNA-DNA duplexes are more sensitive to mismatches, suggesting that a DNA- guided nuclease may not bind as readily to off-target sequences, making them comparatively more specific than RNA-guided nucleases.
  • the guide RNAs usable in the methods described herein can be hybrids, i.e., wherein one or more deoxyribonucleotides, e.g., a short DNA oligonucleotide, replaces all or part of the gRNA, e.g., all or part of the complementarity region of a gRNA.
  • This DNA-based molecule could replace either all or part of the gRNA in a single gRNA system or alternatively might replace all of part of the crRNA and/or tracrRNA in a dual crRNA/tracrRNA system.
  • complementarity region should more reliably target the intended genomic DNA sequences due to the general intolerance of DNA-DNA duplexes to mismatching compared to R A-DNA duplexes.
  • Methods for making such duplexes are known in the art, See, e.g., Barker et al, BMC Genomics. 2005 Apr 22;6:57; and Sugimoto et al, Biochemistry. 2000 Sep 19;39(37): 11270-81.
  • one or both can be synthetic and include one or more modified (e.g., locked) nucleotides or deoxyribonucleotides .
  • complexes of Cas9 with these synthetic gR could be used to improve the genome-wide specificity of the CRISPR/Cas9 nuclease system.
  • the methods described can include expressing in a cell, or contacting the cell with, a Cas9 gRNA plus a fusion protein as described herein.
  • the nucleic acid encoding the guide RNA can be cloned into an intermediate vector for transformation into prokaryotic or eukaryotic cells for replication and/or expression.
  • Intermediate vectors are typically prokaryote vectors, e.g., plasmids, or shuttle vectors, or insect vectors, for storage or manipulation of the nucleic acid encoding the fusion proteins for production of the fusion proteins.
  • the nucleic acid encoding the fusion proteins can also be cloned into an expression vector, for administration to a plant cell, animal cell, preferably a mammalian cell or a human cell, fungal cell, bacterial cell, or protozoan cell.
  • a sequence encoding a fusion protein is typically subcloned into an expression vector that contains a promoter to direct transcription.
  • Suitable bacterial and eukaryotic promoters are well known in the art and described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual (3d ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al, eds., 2010).
  • Bacterial expression systems for expressing the engineered protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al, 1983, Gene 22:229-235).
  • Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available.
  • the promoter used to direct expression of a nucleic acid depends on the particular application. For example, a strong constitutive promoter is typically used for expression and purification of fusion proteins.
  • a constitutive or an inducible promoter can be used, depending on the particular use of the guide RNA.
  • a preferred promoter for administration of the guide RNA can be a weak promoter, such as HSV TK or a promoter having similar activity.
  • the promoter can also include elements that are responsive to transactivation, e.g., hypoxia response elements, Gal4 response elements, lac repressor response element, and small molecule control systems such as tetracycline-regulated systems and the RU-486 system (see, e.g., Gossen & Bujard, 1992, Proc. Natl. Acad. Sci. USA, 89:5547; Oligino et al, 1998, Gene Ther., 5:491-496; Wang et al, 1997, Gene Ther., 4:432-441; Neering et al, 1996, Blood, 88: 1147-55; and Rendahl et al, 1998, Nat. BiotechnoL, 16:757-761).
  • elements that are responsive to transactivation e.g., hypoxia response elements, Gal4 response elements, lac repressor response element, and small molecule control systems such as tetracycline-regulated systems and the RU-486 system (see, e
  • the expression vector typically contains a transcription unit or expression cassette that contains all the additional elements required for the expression of the nucleic acid in host cells, either prokaryotic or eukaryotic.
  • Atypical expression cassette thus contains a promoter operably linked, e.g., to the nucleic acid sequence encoding the gRNA, and any signals required, e.g., for efficient polyadenylation of the transcript, transcriptional termination, ribosome binding sites, or translation termination.
  • Additional elements of the cassette may include, e.g., enhancers, and heterologous spliced intronic signals.
  • the particular expression vector used to transport the genetic information into the cell is selected with regard to the intended use of the gRNA, e.g., expression in plants, animals, bacteria, fungus, protozoa, etc.
  • Standard bacterial expression vectors include plasmids such as pBR322 based plasmids, pSKF, pET23D, and commercially available tag-fusion expression systems such as GST and LacZ.
  • Expression vectors containing regulatory elements from eukaryotic viruses are often used in eukaryotic expression vectors, e.g., SV40 vectors, papilloma virus vectors, and vectors derived from Epstein-Barr virus.
  • eukaryotic vectors include pMSG, pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 late promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
  • the vectors for expressing the guide RNAs can include RNA Pol III promoters to drive expression of the guide RNAs, e.g., the HI, U6 or 7SK promoters. These human promoters allow for expression of gRNAs in mammalian cells following plasmid transfection. Alternatively, a T7 promoter may be used, e.g., for in vitro transcription, and the RNA can be transcribed in vitro and purified. Vectors suitable for the expression of short RNAs, e.g., siRNAs, shRNAs, or other small RNAs, can be used. With the Cys4-based multiplex system described in Figure 4B, multiple gRNAs can be expressed in a single transcript (driven by a RNA Pol II or Pol III promoter) and then cleaved out from that larger transcript.
  • RNA Pol III promoters to drive expression of the guide RNAs.
  • Some expression systems have markers for selection of stably transfected cell lines such as thymidine kinase, hygromycin B phosphotransferase, and dihydrofolate reductase.
  • High yield expression systems are also suitable, such as using a baculovirus vector in insect cells, with the gRNA encoding sequence under the direction of the polyhedrin promoter or other strong baculovirus promoters.
  • the elements that are typically included in expression vectors also include a replicon that functions in E. coli, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of recombinant sequences.
  • Standard transfection methods are used to produce bacterial, mammalian, yeast or insect cell lines that express large quantities of protein, which are then purified using standard techniques (see, e.g., Colley et al, 1989, J. Biol. Chem., 264: 17619-22; Guide to Protein Purification, in Methods in Enzymology, vol. 182 (Deutscher, ed., 1990)). Transformation of eukaryotic and prokaryotic cells are performed according to standard techniques (see, e.g., Morrison, 1977, J. Bacteriol. 132:349-351; Clark-Curtiss & Curtiss, Methods in Enzymology 101 :347-362 (Wu et al, eds, 1983).
  • Any of the known procedures for introducing foreign nucleotide sequences into host cells may be used. These include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, nucleofection, liposomes,
  • microinjection naked DNA, plasmid vectors, viral vectors, both episomal and integrative, and any of the other well-known methods for introducing cloned genomic DNA, cDNA, synthetic DNA or other foreign genetic material into a host cell (see, e.g., Sambrook et al., supra). It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing at least one gene into the host cell capable of expressing the gRNA.
  • the present invention includes the vectors and cells comprising the vectors.
  • RGNs CRISPR RNA-guided nucleases
  • Example 1 The following materials and methods were used in Example 1.
  • DNA oligonucleotides harboring variable 20 nt sequences for Cas9 targeting were annealed to generate short double-strand DNA fragments with 4 bp overhangs compatible with ligation into BsmBI-digested plasmid pMLM3636. Cloning of these annealed oligonucleotides generates plasmids encoding a chimeric +103 single-chain guide RNA with 20 variable 5 ' nucleotides under expression of a U6 promoter
  • pMLM3636 and the expression plasmid pJDS246 (encoding a codon optimized version of Cas9) used in this study are both available through the non-profit plasmid distribution service Addgene (addgene.org/crispr-cas).
  • U20S.EGFP cells harboring a single integrated copy of an EGFP-PEST fusion gene were cultured as previously described (Reyon et al, Nat Biotech 30, 460- 465 (2012)).
  • 200,000 cells were Nucleofected with the indicated amounts of gRNA expression plasmid and pJDS246 together with 30 ng of a Td- tomato-encoding plasmid using the SE Cell Line 4D-NucleofectorTM X Kit (Lonza) according to the manufacturer's protocol. Cells were analyzed 2 days post- transfection using a BD LSRII flow cytometer. Transfections for optimizing gR A/Cas9 plasmid concentration were performed in triplicate and all other transfections were performed in duplicate.
  • PCR reactions were performed using Phusion Hot Start II high-fidelity DNA polymerase (NEB). Most loci amplified successfully using touchdown PCR (98 °C, 10 s; 72-62 °C, -1 °C/cycle, 15 s; 72 °C, 30 s]10 cycles, [98 °C, 10 s; 62 °C, 15 s; 72 °C, 30 s]25 cycles). PCR for the remaining targets were performed with 35 cycles at a constant annealing temperature of 68 °C or 72 °C and 3% DMSO or 1M betaine, if necessary. PCR products were analyzed on a QIAXCEL capillary electrophoresis system to verify both size and purity. Validated products were treated with ExoSap-IT (Affymetrix) and sequenced by the Sanger method (MGH DNA Sequencing Core) to verify each target site.
  • NEB Phusion Hot Start II high-fidelity DNA polymerase
  • Lipofectamine LTX reagent according to the manufacturer's instructions (Life Technologies). Genomic DNA was harvested from transfected U20S.EGFP,
  • HEK293, or K562 cells using the QIAamp DNA Blood Mini Kit (QIAGEN), according to the manufacturer's instructions.
  • QIAGEN QIAamp DNA Blood Mini Kit
  • PCR was then performed using these genomic DNAs as templates as described above and purified using Ampure XP beads (Agencourt) according to the manufacturer's instructions. T7EI assays were performed as previously described (Reyon et al, 2012, supra).
  • EGFP enhanced green fluorescent protein
  • the activities of nucleases targeted to a single integrated EGFP reporter gene can be quantified by assessing loss of fiuorescence signal in human U20S.EGFP cells caused by inactivating frameshift insertion/deletion (indel) mutations introduced by error prone non-homologous end-joining (NHEJ) repair of nuclease-induced double- stranded breaks (DSBs) (Fig. 2B).
  • sgRNAs three -100 nt single gRNAs targeted to different sequences within EGFP were used, as follows:
  • EGFP Site 1 GGGCACGGGCAGCTTGCCGGTGG (SEQ ID NO : 1 )
  • EGFP Site 2 GATGCCGTTCTTCTGCTTGTCGG (SEQ ID NO:2)
  • Each of these sgRNAs can efficiently direct Cas9-mediated disruption of EGFP expression (see Example le and 2a, and FIGs. 3E (top) and 3F (top)).
  • variant sgRNAs were generated for each of the three target sites harboring Watson-Crick transversion mismatches at positions 1 through 19 (numbered 1 to 20 in the 3' to 5' direction; see Fig. 1) and the abilities of these various sgR As to direct Cas9-mediated EGFP disruption in human cells tested (variant sgRNAs bearing a substitution at position 20 were not generated because this nucleotide is part of the U6 promoter sequence and therefore must remain a guanine to avoid affecting expression.)
  • target site #1 was particularly sensitive to a mismatch at position 2 whereas target site #3 was most sensitive to mismatches at positions 1 and 8.
  • variant sgRNAs were constructed bearing increasing numbers of mismatched positions ranging from positions 19 to 15 in the 5' end of the gRNA targeting region (where single and double mismatches appeared to be better tolerated).
  • sgRNAs that target three different sites in the VEGFA gene, one in the EMX1 gene, one in the RNF2 gene, and one in the FANCF gene were used. These six sgRNAs efficiently directed Cas9-mediated indels at their respective endogenous loci in human U20S.EGFP cells as detected by T7
  • T7EI Endonuclease I assay
  • U20S.EGFP cells The loci assessed included all genomic sites that differ by one or two nucleotides as well as subsets of genomic sites that differ by three to six nucleotides and with a bias toward those that had one or more of these mismatches in the 5' half of the gRNA targeting sequence.
  • T7EI assay four off-target sites (out of 53 candidate sites examined) for VEGFA site 1, twelve (out of 46 examined) for VEGFA site 2, seven (out of 64 examined) for VEGFA site 3 and one (out of 46 examined) for the EMX1 site were readily identified. No off-target mutations were detected among the 43 and 50 potential sites examined for the RNF2 or FANCF genes, respectively.
  • the rates of mutation at verified off-target sites were very high, ranging from 5.6% to 125% (mean of 40%) of the rate observed at the intended target site.
  • These bona fide off-targets included sequences with mismatches in the 3' end of the target site and with as many as a total of five mismatches, with most off-target sites occurring within protein coding genes.
  • DNA sequencing of a subset of off-target sites provided additional molecular confirmation that indel mutations occur at the expected RGN cleavage site (Figs. 8A-C).
  • NHEJ-mediated indel mutations at their intended on-target site in these two additional human cell lines (as assessed by T7EI assay), albeit with somewhat lower mutation frequencies than those observed in U20S.EGFP cells.
  • Assessment of the 24 off-target sites for these four RGNs originally identified in U20S.EGFP cells revealed that many were again mutated in HEK293 and K562 cells with frequencies similar to those at their corresponding on-target site.
  • DNA sequencing of a subset of these off-target sites from HEK293 cells provided additional molecular evidence that alterations are occurring at the expected genomic loci.
  • Single guide RNAs were generated for three different sequences (EGFP SITES 1-3, shown above) located upstream of EGFP nucleotide 502, a position at which the introduction of frameshift mutations via non-homologous end- joining can robustly disrupt expression of EGFP (Maeder, M.L. et al, Mol Cell 31, 294-301 (2008); Reyon, D. et al, Nat Biotech 30, 460-465 (2012)).
  • gRNA-expressing plasmid amounts (12.5 to 250 ng) was initially trans fected together with 750 ng of a plasmid expressing a codon-optimized version of the Cas9 nuclease into our U20S.EGFP reporter cells bearing a single copy, constitutively expressed EGFP-PEST reporter gene. All three RGNs efficiently disrupted EGFP expression at the highest concentration of gRNA plasmid (250 ng) (Fig. 3E (top)).
  • RGNs for target sites #1 and #3 exhibited equivalent levels of disruption when lower amounts of gRNA-expressing plasmid were transfected whereas RGN activity at target site #2 dropped immediately when the amount of gRNA-expressing plasmid transfected was decreased (Fig. 3E (top)).
  • the amount of Cas9-encoding plasmid (range from 50 ng to 750 ng) transfected into our U20S.EGFP reporter cells was titrated EGFP disruption assayed. As shown in Fig. 3F (top), target site #1 tolerated a three-fold decrease in the amount of Cas9-encoding plasmid transfected without substantial loss of EGFP disruption activity. However, the activities of RGNs targeting target sites #2 and #3 decreased immediately with a three-fold reduction in the amount of Cas9 plasmid transfected (Fig. 3F (top)).
  • Off-target sites for each of the six RGNs targeted to the VEGFA, RNF2, FANCF, and EMX1 genes and the three RGNs targeted to EGFP Target Sites #1, #2 and #3 were identified in human genome sequence build GRCh37. Mismatches were only allowed for the 20 nt region to which the gRNA anneals and not to the PAM sequence.
  • Example 2 Using pairs of guideRNAs with FokI-dCas9 fusion proteins
  • Monomeric CRISPR-Cas9 nucleases are widely used for targeted genome editing but can induce unwanted off-target mutations with high frequencies.
  • This example describes new dimeric RNA-guided Fokl Nucleases (RFNs) that recognize an extended, double-length sequence and that strictly depend on two single guide RNAs (gRNAs) for cleavage activity. RFNs can robustly edit DNA sequences in endogenous human genes with high efficiencies. Additionally, a method for expressing gRNAs bearing any 5 ' end nucleotide is described, a critical advance that gives dimeric RFNs a useful targeting range.
  • RFNs RNA-guided Fokl Nucleases
  • monomeric Cas9 nickases In direct comparisons, monomeric Cas9 nickases generally induce unwanted indels and unexpected focal point mutations with higher frequencies than RFNs directed by a matched single gRNA.
  • RFNs combine the ease of CRISPR RNA-based targeting with the specificity enhancements of dimerization and provide an important new platform for research and therapeutic applications that require highly precise genome editing.
  • Plasmids encoding single or multiplex gRNAs were assembled in a single-step ligation of annealed target site oligosduplexes (Integrated DNA Technologies) and a constant region oligoduplex (for multiplex gRNAs) with BsmBI-digested Csy4- flanked gRNA backbone (pSQT1313; Addgene).
  • Multiplex gRNA encoding plasmids were constructed by ligating: 1) annealed oligos encoding the first target site, 2) phosphorylated annealed oligos encoding crRNA, tracrRNA, and Csy4-binding site, and 3) annealed oligos encoding the second target site, into a U6-Csy4site-gRNA plasmid backbone digested with BsmBI Type lis restriction enzyme.
  • Csy4 RNA binding sites were attached to the 3' and 5' ends of a gRNA sequence and expressed with Cas9 in cells.
  • the Csy4 RNA binding site sequence ' GUUC ACUGCCGUAUAGGC AGCUAAGAAA' was fused to the 5' and 3' end of the standard gRNA sequence.
  • This sequence is a multiplex gRNA sequence flanked by Csy4 sites (underlined). Functionally, encoding these in multiplex on one transcript should have the same result as encoding them separately. Although all pairs of Csy4-flanked sgRNAs were expressed in a multiplex context in the experiments described herein, the sgRNAs can be encoded in multiplex sgRNAs separated by Csy4 sites encoded on one transcript as well as individual sgRNAs that have an additional Csy4 sequence.
  • the first N20 sequence represents the sequence complementary to one strand of the target genomic sequence
  • the second N20 sequence represents the sequence complementary to the other strand of the target genomic sequence.
  • a plasmid encoding the Csy4 recognition site containing gR A was co- transfected with plasmid encoding Cas9 and Csy4 proteins separated by a '2A' peptide linkage.
  • the results showed that gR As with Csy4 sites fused to the 5' and 3 ' ends remained capable of directing Cas9-mediated cleavage in human cells using the U20S-EGFP disruption assay previously described.
  • Csy4 R A binding sites can be attached to 3 ' end of a gRNA sequence and complexes of these Csy4 site- containing gRNAs with Cas9 remain functional in the cell.
  • the sequences of the FokI-dCas9 fusions are shown below, and include a GGGGS (SEQ ID NO:23) linker (underlined) between the Fokl and dCas9 and a nuclear localization sequence.
  • FokI-dCas9 amino acid sequence (FokI-G4S-dCas9-nls-3XFLAG) MQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNSTQDRILEMKVMEFFMKVYGYR GKHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMQRYVEENQTRN KHINPNEWWKVYPSSVTEFKFLFVSGHFKGNYKAQLTRLNHITNCNGAVLSVEELLI GGEMIKAGTLTLEEVRRKFNNGEINFGGGGSDKKYS IGLAIGTNSVGWAVITDEYKV PSKKFKVLGN DRHS IKKNLIGALLFDSGE AEATRLKRTARRRYTRRKNRICYLQE IFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFG IVDEVAYHEKYP IYHLRKK LVDS DKADLRLIYLALAHMI
  • FokI-dCas9 nucleotide sequence (FokI-G4S-dCas9-nls-3XFLAG)
  • U20S.EGFP cells were cultured in the presence of 400 ⁇ g/ml of G418.
  • U20S cells and U20S.EGFP cells were transfected using the DN-100 program of a Lonza 4D-Nucleofector according to the manufacturer's instructions.
  • 750 ng of pCAG-Csy4-FokI-dCas9-nls nuclease plasmid and 250 ng of gRNA encoding plasmids were transfected together with 50 ng tdTomato expression plasmid (Clontech) as a transfection control.
  • tdTomato expression plasmid (Clontech)
  • U20S.EGFP cells 975 ng of human codon optimized pCAG-Csy4-T2A-nls-hFokI- dCas9-nls (SQT1601) or pCAG-Cas9-D10A nickase (NW3) were transfected along with 325 ng of gRNA vector and 10 ng of Td tomato expression plasmid and analyzed 3 days after transfection.
  • HEK293 cells were transfected with 750 ng of nuclease plasmid, 250 ng of gRNA expression plasmid and 10 ng of Td tomato, using
  • the EGFP disruption assay was performed as previously described (see Example 1 and Reyon et al, Nat Biotech 30, 460-465 (2012)) using U20S.EGFP reporter cells. Cells were assayed for EGFP and tdTomato expression using an BD Biosciences LSR II or Fortessa FACS analyzer.
  • T7E1 assays were performed as previously described (Reyon et al, Nat Biotech 30, 460-465 (2012)). Briefly, genomic DNA was isolated 72 hours post transfection using the Agencourt DNAdvance Genomic DNA Isolation kit (Beckman Coulter Genomics) according to the manufacturer's instructions with a Sciclone G3 liquid-handling workstation (Caliper). PCR reactions to amplify genomic loci were performed using Phusion Hot- start Flex DNA polymerase (New England Biolabs).
  • Samples were amplified using a two-step protocol (98 °C, 30 sec; (98 °C, 7 sec; 72 °C, 30 sec) x 35; 72 °C, 5 min) or a touchdown PCR protocol ((98 °C, 10 s; 72-62 °C, -1 °C/cycle, 15 s; 72 °C, 30 s) 10 cycles, (98 °C, 10 s; 62 °C, 15 s; 72 °C, 30 s) 25 cycles).
  • 200 ng of purified PCR amplicons were denatured, hybridized, and treated with T7 Endonuclease I (New England Biolabs). Mutation frequency was quantified using a Qiaxcel capillary electrophoresis instrument (Qiagen) as previously described (Reyon et al, Nat Biotech 30, 460-465 (2012)).
  • Short 200-350 bp PCR products were amplified using Phusion Hot- start FLEX DNA polymerase. PCR products were purified using Ampure XP beads (Beckman Coulter Genomics) according to manufacturer's instructions. Dual-indexed TruSeq Illumina deep sequencing libraries were prepared using a high-throughput library preparation system (Kapa Biosystems) on a Sciclone G3 liquid-handling workstation. Final adapter-ligated libraries were quantified using a Qiaxcel capillary electrophoresis instrument (Qiagen). 150 bp paired end sequencing was performed on an Illumina MiSeq Sequencer by the Dana-Farber Cancer Institute Molecular Biology Core.
  • MiSeq paired-end reads were mapped to human genome reference GChr37 using bwa. Reads with an average quality score >30 were analyzed for insertion or deletion mutations that overlapped the intended target or candidate off-target nuclease binding site. Mutation analyses were conducted using the Genome Analysis Toolkit (GATK) and Python.
  • GATK Genome Analysis Toolkit
  • a target-site matching algorithm was implemented that looks for matches with less than a specified number of mismatches in a sliding window across the human genome.
  • Example 2a Rationale for designing dimeric RNA-guided nucleases
  • Example 2b Multiplex expression of gRNAs without 5 '-end nucleotide limitations
  • the targeting range for a dimeric RNA-guided nuclease would be low using existing gRNA expression methods.
  • Two sequence requirements typically restrict the targeting range of a dCas9 monomer: the requirement for a PAM sequence of 5'- NGG that is specified by the dCas9 and a requirement for a G nucleotide at the 5' end of the gRNA imposed by the use of a U6 promoter in most expression vectors. If, however, the requirement for the 5 ' G in the gRNA could be relieved, then the targeting range would improve by 16-fold.
  • a plasmid was constructed from which two gRNAs, each flanked by cleavage sites for the Csy4 ribonuclease (Haurwitz et al, Science 329, 1355-1358 (2010)), can be expressed within a single RNA transcribed from a U6 promoter (Fig. 4B). Csy4 would be expected to process this transcript thereby releasing the two gRNAs.
  • each processed gRNA should retain a Csy4 recognition site on its 3' end with a Csy4 protein bound to that site (Fig. 4B).
  • gRNAs with any 5' nucleotide. This system was tested by using it to express two gRNAs targeted to sites within the EGFP reporter gene.
  • Example 2c Construction and optimization of dimeric RNA-guided nucleases Two different hybrid proteins harboring the Fokl nuclease domain and the dCas9 protein were constructed: one in which the Fokl nuclease domain is fused to the carboxy-terminus of dCas9 (dCas9-FokI) and the other in which it is fused to the amino-terminus (FokI-dCas9) (Fig. 5A).
  • the dCas9-FokI protein is analogous in architecture to ZFNs and TALENs (Fig. 5A).
  • the dCas9-FokI protein did not show detectable EGFP disruption activity when co-expressed with any of the 60 gRNA pairs in human U20S.EGFP cells (Fig. 5E).
  • screening of the FokI-dCas9 protein with the same 60 gRNA pairs did reveal EGFP disruption activity on target sites composed of half-sites in the PAM out orientation and with spacer lengths of 13 to 17 bps and of 26 bps (approximately one turn of the DNA helix more than the 13-17 bp spacer lengths) (Fig. 5B).
  • FokI-dCas9 can be directed by two appropriately positioned gRNAs to efficiently cleave a full-length target site of interest.
  • the complex of two FokI-dCas9 fusions and two gRNAs are referred to herein as RNA-guided Fokl Nucleases (RFNs).
  • gRNA pairs were designed for 12 different target sites in nine different human genes (Table 2). Eleven of the 12 RFNs tested introduced indels with high efficiencies (range of 3 to 40%) at their intended target sites in human U20S.EGFP cells as judged by T7EI assay (Table 2). Similar results were obtained with these same 12 RFN pairs in HEK293 cells (Table 2). Sanger sequencing of successfully targeted alleles from U20S.EGFP cells revealed the introduction of a range of indels (primarily deletions) at the expected cleavage site (Fig. 5F). The high success rate and high efficiencies of modifications observed in two different human cell lines demonstrate the robustness of RFNs for modifying endogenous human genes.
  • Example 2d RFNs possess extended specificities for their cleavage sites To test whether RFNs possess enhanced recognition specificities associated with dimerization, whether these nucleases strictly depend upon the presence of both gRNAs in a pair was examined. In an ideal dimeric system, single gRNAs should not be able to efficiently direct FokI-dCas9-induced indels. To perform an initial test, two pairs of gRNAs directed to two target sites in EGFP were used that had been shown to efficiently direct FokI-dCas9-induced indels to their target sites (EGFP sites 47 and 81) in human U20S.EGFP cells (Fig. 5C).
  • Example 2e Monomeric Cas9 nickases induce higher rates of mutagenesis than single gRNA/FokI-dCas9 complexes
  • FokI-dCas9 and Cas9 nickase were compared in the presence of a single gRNA at six dimeric human gene target sites (a total of 12 half-sites; Table 4). These particular sites were chosen because monomeric Cas9 nickases directed by just one and/or the other gRNA in a pair could induce indel mutations at these targets.
  • Table 4 the activities of FokI-dCas9 or Cas9 nickase were assessed in the presence of both or only one or the other gRNAs.
  • Example 2f Dimeric RFNs possess a high degree of specificity
  • Dimeric RFNs directed by two gRNAs are not expected to induce appreciable off-target mutations in human cells.
  • RFNs directed by a pair of gRNAs to cleave a full-length sequence composed of two half-sites, would be expected to specify up to 44 bps of DNA in the target site. A sequence of this length will, by chance, almost always be unique (except in certain circumstances where the target might lie in duplicated genome sequence).
  • the most closely matched sites in the genome to this full-length site should, in most cases, possess a large number of mismatches, which in turn would be expected to minimize or abolish cleavage activity by an RFN dimer.
  • VEGFA-2 1 4 9 99 447 1675 5608 18599
  • VEGFA-3 1 20 120 623 2783 References

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

Many studies have shown that CRISPR-Cas nucleases can tolerate up to five mismatches and still cleave; it is hard to predict the effects of any given single or combination of mismatches on activity. Taken together, these nucleases can show significant off-target effects but it can be challenging to predict these sites. Described herein are methods for increasing the specificity of genome editing using the CRISPR/Cas system, e.g., using RNA-guided Foki Nucleases (RFNs), e.g., Fokl- Cas9 or Foki-dCas9-based fusion proteins.

Description

Using RNA-guided Fokl Nucleases (RFNs) to Increase Specificity for RNA-Guided Genome Editing
CLAIM OF PRIORITY
This application claims priority under 35 USC § 119(e) to U.S. Patent
Application Serial Nos. 61/838,178, filed on June 21, 2013; 61/838,148, filed on June 21, 2013; 61/921,007, filed on December 26, 2013; and PCT/US2014/028630, filed March 14, 2014. The entire contents of the foregoing are hereby incorporated by reference.
FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
This invention was made with Government support under Grant Nos. DPI GM 105378 awarded by the National Institutes of Health. The Government has certain rights in the invention.
TECHNICAL FIELD
Methods for increasing specificity of RNA-guided genome editing, e.g., editing using CRISPR/Cas9 systems, using RNA-guided Fokl Nucleases (RFNs), e.g., FokI-dCas9 fusion proteins.
BACKGROUND
Recent work has demonstrated that clustered, regularly interspaced, short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems (Wiedenheft et al, Nature 482, 331-338 (2012); Horvath et al, Science 327, 167-170 (2010); Terns et al, Curr Opin Microbiol 14, 321-327 (2011)) can serve as the basis genome editing in bacteria, yeast and human cells, as well as in vivo in whole organisms such as fruit flies, zebrafish and mice (Wang et al, Cell 153, 910-918 (2013); Shen et al, Cell Res (2013); Dicarlo et al, Nucleic Acids Res (2013); Jiang et al, Nat Biotechnol 31 , 233- 239 (2013); Jinek et al, Elife 2, e00471 (2013); Hwang et al, Nat Biotechnol 31 , 227- 229 (2013); Cong et al, Science 339, 819-823 (2013); Mali et al, Science 339, 823- 826 (2013c); Cho et al, Nat Biotechnol 31 , 230-232 (2013); Gratz et al, Genetics 194(4): 1029-35 (2013)). The Cas9 nuclease from S. pyogenes (hereafter simply Cas9) can be guided via base pair complementarity between the first 20 nucleotides of an engineered gRNA and the complementary strand of a target genomic DNA sequence of interest that lies next to a protospacer adjacent motif (PAM), e.g., a PAM matching the sequence NGG or NAG (Shen et al, Cell Res (2013); Dicarlo et al, Nucleic Acids Res (2013); Jiang et al, Nat Biotechnol 31 , 233-239 (2013); Jinek et al, Elife 2, e00471 (2013); Hwang et al, Nat Biotechnol 31, 227-229 (2013); Cong et al, Science 339, 819-823 (2013); Mali et al, Science 339, 823-826 (2013c); Cho et al, Nat
Biotechnol 31, 230-232 (2013); Jinek et al, Science 337, 816-821 (2012)). Previous studies performed in vitro (Jinek et al, Science 337, 816-821 (2012)), in bacteria (Jiang et al, Nat Biotechnol 31, 233-239 (2013)) and in human cells (Cong et al., Science 339, 819-823 (2013)) have shown that Cas9-mediated cleavage can, in some cases, be abolished by single mismatches at the gRNA/target site interface, particularly in the last 10-12 nucleotides (nts) located in the 3' end of the 20 nucleotide (nt) gRNA complementarity region.
SUMMARY
Many studies have shown that CRISPR-Cas nucleases can tolerate up to five mismatches and still cleave; it is hard to predict the effects of any given single or combination of mismatches on activity. Taken together, these nucleases can show significant off-target effects but it can be challenging to predict these sites. Described herein are methods for increasing the specificity of genome editing using the
CRISPR/Cas system, e.g., using RNA-guided Fokl Nucleases (RFNs), e.g., Fokl- Cas9 or FokI-dCas9-based fusion proteins.
In a first aspect, the invention provides FokI-dCas9 fusion proteins, comprising a Fokl catalytic domain sequence fused to the terminus, e.g., the N terminus, of dCas9, optionally with an intervening linker, e.g., a linker of from 2-30 amino acids, e.g., 4-12 amino acids, e.g., Gly4Ser. In some embodiments, the Fokl catalytic domain comprises amino acids 388-583 or 408-583 of SEQ ID NO:4. In some embodiments, the dCas9 comprises mutations at the dCas9 comprises mutations at D10, E762, H983, or D986; and at H840 or N863; e.g., at: (i) DIOA or DION; and (ii) H840A, H840Y or H840N.
In another aspect, the invention provides nucleic acids encoding these fusion proteins, vector comprising the nucleic acids, and host cells harboring or expressing the nucleic acids, vectors, or fusion proteins. In another aspect, the invention provides methods for inducing a sequence- specific break in a double-stranded DNA molecule, e.g., in a genomic sequence in a cell, the method comprising expressing in the cell, or contacting the cell with, the FokI-dCas9 fusion protein described herein, and:
(a) two single guide R As, wherein each of the two single guide R As include sequences that are each complementary to one strand of the target sequence such that using both guide RNAs results in targeting both strands (i.e., one single guide RNA targets a first strand, and the other guide RNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double-stranded break, or
(b) a tracrRNA and two crRNAs wherein each of the two crRNAs include sequences that are complementary to one strand of the target sequence such that using both crRNAs results in targeting both strands (i.e., one crRNA targets a first strand, and the other crRNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double- stranded break.
In another aspect, the invention provides methods for increasing specificity of RNA-guided genome editing in a cell, the method comprising contacting the cell with an RNA-guided Fokl Nuclease (RFN) fusion protein described herein.
The method may further comprise expressing in the cell, or contacting the cell with, (a) two single guide RNAs, wherein each of the two single guide RNAs include sequences that are each complementary to one strand of the target sequence such that using both guide RNAs results in targeting both strands (i.e., one single guide RNA targets a first strand, and the other guide RNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double-stranded break, or
(b) a tracrRNA and two crRNAs wherein each of the two crRNAs include sequences that are complementary to one strand of the target sequence such that using both crRNAs results in targeting both strands (i.e., one crRNA targets a first strand, and the other crRNA targets the complementary strand), and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double- stranded break. In some embodiments, the two target genomic sequences (i.e., the sequences to which the target complementarity regions of the crR A or single guide R As are complementary) are spaced 10-20 base pairs apart, preferably 13-17 base pairs apart.
In some embodiments, an indel mutation is induced between the two target sequences.
In some embodiments, the specificity of RNA-guided genome editing in a cell is increased.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.
Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.
DESCRIPTION OF DRAWINGS
Figure 1 : Schematic illustrating a gR A/Cas9 nuclease complex bound to its target DNA site. Scissors indicate approximate cleavage points of the Cas9 nuclease on the genomic DNA target site. Note the numbering of nucleotides on the guide RNA proceeds in an inverse fashion from 5' to 3'.
Figure 2A: Schematic illustrating the rationale for truncating the 5' complementarity region of a gRNA. Thick grey lines = target DNA site, thin dark grey line structure = gRNA, grey oval = Cas9 nuclease, black lines indicate base pairing between gRNA and target DNA site.
Figure 2B: Schematic overview of the EGFP disruption assay. Repair of targeted Cas9-mediated double-stranded breaks in a single integrated EGFP-PEST reporter gene by error-prone NHEJ-mediated repair leads to frame-shift mutations that disrupt the coding sequence and associated loss of fluorescence in cells. Figures 2C-F: Activities of RGNs harboring sgRNAs bearing (C) single mismatches, (D) adjacent double mismatches, (E) variably spaced double mismatches, and (F) increasing numbers of adjacent mismatches assayed on three different target sites in the EGFP reporter gene sequence. Mean activities of replicates (see Online Methods) are shown, normalized to the activity of a perfectly matched gRNA. Error bars indicate standard errors of the mean. Positions mismatched in each gRNA are highlighted in grey in the grid below. Sequences of the three EGFP target sites were as follows:
EGFP Site 1 GGGCACGGGCAGCTTGCCGGTGG (SEQ ID NO : 1 ) EGFP Site 2 GATGCCGTTCTTCTGCTTGTCGG (SEQ ID NO:2)
EGFP Site 3 GGTGGTGCAGATGAACTTCAGGG (SEQ ID NO:3) Figure 2G: Mismatches at the 5' end of the gRNA make CRISPR/Cas more sensitive more 3' mismatches. The gRNAs Watson-Crick base pair between the RNA&DNA with the exception of positions indicated with an "m" which are mismatched using the Watson-Crick transversion (i.e. EGFP Site#2 M18-19 is mismatched by changing the gRNA to its Watson-Crick partner at positions 18 & 19. Although positions near the 5 ' of the gRNA are generally very well tolerated, matches in these positions are important for nuclease activity when other residues are mismatched. When all four positions are mismatched, nuclease activity is no longer detectable. This further demonstrates that matches at these 5' position can help compensate for mismatches at other more 3' positions. Note these experiments were performed with a non-codon optimized version of Cas9 which can show lower absolute levels of nuclease activity as compared to the codon optimized version.
Figure 2H: Efficiency of Cas9 nuclease activities directed by gRNAs bearing variable length complementarity regions ranging from 15 to 25 nts in a human cell- based U20S EGFP disruption assay. Expression of a gRNA from the U6 promoter requires the presence of a 5 ' G and therefore it was only possible to evaluate gRNAs harboring certain lengths of complementarity to the target DNA site (15, 17, 19, 20, 21, 23, and 25 nts).
Figure 3 A: Efficiencies of EGFP disruption in human cells mediated by Cas9 and full-length or shortened gRNAs for four target sites in the EGFP reporter gene. Lengths of complementarity regions and corresponding target DNA sites are shown. Ctrl = control gRNA lacking a complementarity region. Figure 3B: Efficiencies of targeted indel mutations introduced at seven different human endogenous gene targets by matched standard and tru-RGNs.
Lengths of complementarity regions and corresponding target DNA sites are shown. Indel frequencies were measured by T7EI assay. Ctrl = control gRNA lacking a complementarity region.
Figure 3C: DNA sequences of indel mutations induced by RGNs using a tru- gRNA or a matched full-length gRNA targeted to the EMXl site. The portion of the target DNA site that interacts with the gRNA complementarity region is highlighted in grey with the first base of the PAM sequence shown in lowercase. Deletions are indicated by dashes highlighted in grey and insertions by italicized letters highlighted in grey. The net number of bases deleted or inserted and the number of times each sequence was isolated are shown to the right.
Figure 3D: Efficiencies of precise HDR/ssODN-mediated alterations introduced at two endogenous human genes by matched standard and tru-RGNs. %HDR was measured using a BamHl restriction digest assay (see the Experimental Procedures for Example 2). Control gRNA = empty U6 promoter vector.
Figure 3E: U20S.EGFP cells were transfected with variable amounts of full- length gRNA expression plasmids (top) or tru-gRNA expression plasmids (bottom) together with a fixed amount of Cas9 expression plasmid and then assayed for percentage of cells with decreased EGFP expression. Mean values from duplicate experiments are shown with standard errors of the mean. Note that the data obtained with tru-gRNA matches closely with data from experiments performed with full- length gRNA expression plasmids instead of tru-gRNA plasmids for these three EGFP target sites.
Figure 3F: U20S.EGFP cells were transfected with variable amount of Cas9 expression plasmid together with variable amounts of full-length gRNA expression plasmids (top) or tru-gRNA expression plasmids (bottom) (amounts determined for each tru-gRNA from the experiments of Figure 3E). Mean values from duplicate experiments are shown with standard errors of the mean. Note that the data obtained with tru-gRNA matches closely with data from experiments performed with full- length gRNA expression plasmids instead of tru-gRNA plasmids for these three EGFP target sites. The results of these titrations determined the concentrations of plasmids used in the EGFP disruption assays performed in Examples 1 and 2. Figures 4A-C. RNA-guided Fokl nucleases and a CRISPR/Cas Subtype Ypest protein 4 (Csy4)-based multiplex gRNA expression system.
(a) Schematic overview of RNA-guided Fokl nucleases. Two FokI-dCas9 fusion proteins are recruited to adjacent target sites by two different gRNAs in order to facilitate Fokl dimerization and DNA cleavage.
(b) Schematic overview of a Csy4-based multiplex gRNA expression system. Two gRNAs (with any 5 ' end nucleotide) are co-expressed in a single transcript from a U6 promoter with each gRNA flanked by Csy4 recognition sites. Csy4 cleaves and releases gRNAs from the transcript. The Csy4 recognition site remains at the 3 ' end of the gRNA with a Csy4 nuclease bound to that site.
(c) Validation of the multiplex, Csy4-based system. Two gRNAs targeted to adjacent sites in EGFP were expressed in a single RNA transcript using the Csy4- based system in human U20S.EGFP cells together with Csy4 and Cas9 nucleases. Sequences of indel mutations induced in these cells are shown. The wild-type sequence is shown in the top with both target sites highlighted in grey and PAM sequences shown as underlined text. Deletions are indicated by dashes against gray background and insertions by lowercase letters against a grey background. To the right of each sequence, the sizes of insertions (+) or deletions (Δ) are specified.
Figures 5A-F. Design and optimization of RNA-guided Fokl nucleases.
(a) Schematic illustrations of a ZFN, TALEN, FokI-dCas9 fusion, and dCas9-
Fokl fusion.
(b) Screening the EGFP disruption activities of FokI-dCas9 fusion with gRNA pairs targeted to half-sites in one of two orientations: PAMs in (left panel) and PAMs out (right panel). Half-sites were separated by spacer sequences of variable lengths ranging from 0 to 31 bps. EGFP disruption was quantified by flow cytometry, n = 1. Corresponding data for the dCas9-FokI fusion and the same gRNA pairs is shown in Fig. 5E.
(c) Additional assessment of FokI-dCas9-mediated EGFP disruption activities on target sites with half-sites oriented with their PAMs out and with spacer lengths ranging from 10 to 20 bp. EGFP disruption was quantified by flow cytometry. Error bars indicate standard errors of the mean (s.e.m.), n = 2.
(d) Mean EGFP disruption values of the data from (c) grouped according to spacer length. Error bars represent s.e.m. (e) These plots show the results of a screen for dCas9-FokI activity in EGFP disruption assay in the U20S.EGFP cells with 60 gRNA pairs with spacings of 0-31 bp and PAM in and PAM out orientations.
(f) Sequences of FokI-dCas9 induced mutations in U20S cells are shown. The 23-nt target sequence bound by Cas9 or FokI-dCas9 is labeled in grey. The protospacer adjacent motif or PAM sequence is labeled in boldface with underlining. Deletions are marked with dashes on a light grey background. Insertions are highlighted in grey. The net number of bases inserted or deleted are indicated in a column directly to the right of the sequences.
Figures 6A-D. Dimerization of FokI-dCas9 RFNs is required for efficient genome editing activity.
(a) EGFP disruption activities of two RFN pairs assessed in the presence of correctly targeted gRNA pairs (to EGFP sites 47 and 81) and pairs in which one or the other of the gRNAs has been replaced with another gRNA targeted to a non- EGFP sequence (in the VEGFA gene). EGFP disruption was quantified by flow cytometry. EGFP, Enhanced Green Fluorescent Protein; VEGFA, Vascular
Endothelial Growth Factor A. Error bars represent standard errors of the mean (s.e.m.), n = 3.
(b) Quantification of mutagenesis frequencies by T7EI assay performed with genomic DNA from the same cells used in the EGFP disruption assay of (a). Error bars represent s.e.m., n = 3.
(c) Activities of RFNs targeted to sites in the APC, MLHl and VEGFA genes. For each target, we co-expressed FokI-dCas9 with a pair of cognate gRNAs, only one gRNA for the "left" half-site, or only one gRNA for the "right" half-site. Rates of mutagenesis were measured by T7E1 assay. APC, Adenomatous polyposis coli;
MLHl, mutL homo log 1; VEGFA, Vascular Endothelial Growth Factor A. Error bars represent s.e.m., n = 3.
(d) Mutagenesis frequencies of RFNs targeted to VEGFA site 1 at the on- target site and at five previously known off-target (OT) sites for one of the gRNAs used to target VEGFA site 1. Frequencies of mutation were determined by deep sequencing. Each value reported was determined from a single deep sequencing library prepared from genomic DNA pooled from three independent transfection experiments. The value shown for the on-target VEGFA site 1 (marked with an asterisk) is the same as the one shown in Fig. 4a below and is only shown here for ease of comparison with the values presented in this figure.
Figures 7A-B. Mutagenic activities of a Cas9 nickase or FokI-dCas9 co- expressed with a single gRNA.
(a) Indel mutation frequencies induced by FokI-dCas9 (left bars) or Cas9 nickase (middle bars) in the presence of one or two gRNAs targeted to six different human gene sites. For each gene target, we assessed indel frequencies with both gRNAs, only one gRNA for the "left" half-site, or only the other gRNA for the "right" half-site. Mutation frequencies were determined by deep sequencing. Each indel frequency value reported was determined from a single deep sequencing library prepared from genomic DNA pooled from three independent transfection
experiments. VEGFA, Vascular Endothelial Growth Factor A; DDB2, Damage- Specific DNA Binding Protein 2; FANCF, Fanconi Anemia, Complementation Group F; FES, Feline Sarcoma Oncogene; RUNX 1, Runt-Related Transcription Factor 1.
(b) Data from (a) presented as a fold-reduction in the indel frequency comparing values obtained for each target site with a gRNA pair to each of the single gRNA experiments or to the control experiment (no gRNA and no Cas9 nickase or FokI-dCas9). This fold-reduction was calculated for both FokI-dCas9 (left bars in each pair, lighter grey) and Cas9 nickase (right bars in each pair, darker grey).
Figures 8A-C: Single Cas9 nickases can introduce point mutations with high efficiencies into their target sites.
Frequencies of different point mutations found at each position in half-sites targeted by single gRNAs for (a) VEGFA, (b) FANCF, and (c) RUNX1 gene targets in the presence of FokI-dCas9, Cas9 nickase, or a tdTomato control. Mutation frequencies were determined by deep sequencing. Each point mutation value reported was determined from a single deep sequencing library prepared from genomic DNA pooled from three independent transfection experiments. Note that the genomic DNA used for these experiments was isolated from the same cells analyzed for indel mutations in Figs. 7A-B. VEGFA, Vascular Endothelial Growth Factor A; FANCF, Fanconi Anemia, Complementation Group F; RUNX 1, Runt-Related Transcription Factor 1. DETAILED DESCRIPTION
CRISPR RNA-guided nucleases (RGNs) have rapidly emerged as a facile and efficient platform for genome editing. Although Marraffmi and colleagues (Jiang et al., Nat Biotechnol 31, 233-239 (2013)) recently performed a systematic investigation of Cas9 RGN specificity in bacteria, the specificities of RGNs in human cells have not been extensively defined. Understanding the scope of RGN-mediated off-target effects in human and other eukaryotic cells will be critically essential if these nucleases are to be used widely for research and therapeutic applications. The present inventors have used a human cell-based reporter assay to characterize off-target cleavage of Cas9-based RGNs. Single and double mismatches were tolerated to varying degrees depending on their position along the guide RNA (gRNA)-DNA interface. Off-target alterations induced by four out of six RGNs targeted to endogenous loci in human cells were readily detected by examination of partially mismatched sites. The off-target sites identified harbor up to five mismatches and many are mutagenized with frequencies comparable to (or higher than) those observed at the intended on-target site. Thus RGNs are highly active even with imperfectly matched RNA-DNA interfaces in human cells, a finding that might confound their use in research and therapeutic applications.
The results described herein reveal that predicting the specificity profile of any given RGN is neither simple nor straightforward. The EGFP reporter assay experiments show that single and double mismatches can have variable effects on RGN activity in human cells that do not strictly depend upon their position(s) within the target site. For example, consistent with previously published reports, alterations in the 3 ' half of the gRNA/DNA interface generally have greater effects than those in the 5' half (Jiang et al., Nat Biotechnol 31, 233-239 (2013); Cong et al, Science 339, 819-823 (2013); Jinek et al, Science 337, 816-821 (2012)); however, single and double mutations in the 3 ' end sometimes also appear to be well tolerated whereas double mutations in the 5' end can greatly diminish activities. In addition, the magnitude of these effects for mismatches at any given position(s) appears to be site- dependent. Comprehensive profiling of a large series of RGNs with testing of all possible nucleotide substitutions (beyond the Watson-Crick transversions used in our EGFP reporter experiments) may help provide additional insights into the range of potential off-targets. In this regard, the recently described bacterial cell-based method of Marraffini and colleagues (Jiang et al., Nat Biotechnol 31, 233-239 (2013)) or the in vitro, combinatorial library-based cleavage site-selection methodologies previously applied to ZFNs by Liu and colleagues (Pattanayak et al, Nat Methods 8, 765-770 (2011)) might be useful for generating larger sets of RGN specificity profiles.
Despite these challenges in comprehensively predicting RGN specificities, it was possible to identify bona fide off-targets of RGNs by examining a subset of genomic sites that differed from the on-target site by one to five mismatches. Notably, under conditions of these experiments, the frequencies of RGN-induced mutations at many of these off-target sites were similar to (or higher than) those observed at the intended on-target site, enabling the detection of mutations at these sites using the
T7EI assay (which, as performed in our laboratory, has a reliable detection limit of ~2 to 5% mutation frequency). Because these mutation rates were very high, it was possible to avoid using deep sequencing methods previously required to detect much lower frequency ZFN- and TALEN-induced off-target alterations (Pattanayak et al., Nat Methods 8, 765-770 (2011); Perez et al, Nat Biotechnol 26, 808-816 (2008);
Gabriel et al, Nat Biotechnol 29, 816-823 (2011); Hockemeyer et al, Nat Biotechnol 29, 731-734 (2011)). Analysis of RGN off-target mutagenesis in human cells also confirmed the difficulties of predicting RGN specificities - not all single and double mismatched off-target sites show evidence of mutation whereas some sites with as many as five mismatches can also show alterations. Furthermore, the bona fide off- target sites identified do not exhibit any obvious bias toward transition or transversion differences relative to the intended target sequence.
Although off-target sites were seen for a number of RGNs, identification of these sites was neither comprehensive nor genome -wide in scale. For the six RGNs studied, only a very small subset of the much larger total number of potential off- target sequences in the human genome was examined. Although examining such large numbers of loci for off-target mutations by T7EI assay is neither a practical nor a cost-effective strategy, the use of high-throughput sequencing in future studies might enable the interrogation of larger numbers of candidate off-target sites and provide a more sensitive method for detecting bona fide off-target mutations. For example, such an approach might enable the unveiling of additional off-target sites for the two RGNs for which we failed to uncover any off-target mutations. In addition, an improved understanding both of RGN specificities and of any epigenomic factors (e.g., DNA methylation and chromatin status) that may influence RGN activities in cells might also reduce the number of potential sites that need to be examined and thereby make genome-wide assessments of RGN off-targets more practical and affordable.
A number of strategies can be used to minimize the frequencies of genomic off-target mutations. For example, the specific choice of RGN target site can be optimized; given that off-target sites that differ at up to five positions from the intended target site can be efficiently mutated by RGNs, choosing target sites with minimal numbers of off-target sites as judged by mismatch counting seems unlikely to be effective; thousands of potential off-target sites that differ by four or five positions within the 20 bp RNA:DNA complementarity region will typically exist for any given RGN targeted to a sequence in the human genome. It is also possible that the nucleotide content of the gRNA complementarity region might influence the range of potential off-target effects. For example, high GC-content has been shown to stabilize RNA:DNA hybrids (Sugimoto et al, Biochemistry 34, 1121 1-11216 (1995)) and therefore might also be expected to make gRNA/genomic DNA hybridization more stable and more tolerant to mismatches. Additional experiments with larger numbers of gRNAs will be needed to assess if and how these two parameters (numbers of mismatched sites in the genome and stability of the RNA:DNA hybrid) influence the genome-wide specificities of RGNs. However, it is important to note that even if such predictive parameters can be defined, the effect of implementing such guidelines would be to further restrict the targeting range of RGNs.
One potential general strategy for reducing RGN-induced off-target effects might be to reduce the concentrations of gRNA and Cas9 nuclease expressed in the cell. This idea was tested using the RGNs for VEGFA target sites 2 and 3 in
U20S.EGFP cells; transfecting less gRNA- and Cas9-expressing plasmid decreased the mutation rate at the on-target site but did not appreciably change the relative rates of off-target mutations. Consistent with this, high-level off-target mutagenesis rates were also observed in two other human cell types (HEK293 and K562 cells) even though the absolute rates of on-target mutagenesis are lower than in U20S.EGFP cells. Thus, reducing expression levels of gRNA and Cas9 in cells is not likely to provide a solution for reducing off-target effects. Furthermore, these results also suggest that the high rates of off-target mutagenesis observed in human cells are not caused by overexpression of gRNA and/or Cas9.
The finding that significant off-target mutagenesis can be induced by RGNs in three different human cell types has important implications for broader use of this genome-editing platform. For research applications, the potentially confounding effects of high frequency off-target mutations will need to be considered, particularly for experiments involving either cultured cells or organisms with slow generation times for which the outcrossing of undesired alterations would be challenging. One way to control for such effects might be to utilize multiple RGNs targeted to different DNA sequences to induce the same genomic alteration because off-target effects are not random but instead related to the targeted site. However, for therapeutic applications, these findings clearly indicate that the specificities of RGNs will need to be carefully defined and/or improved if these nucleases are to be used safely in the longer term for treatment of human diseases.
Methods for Improving Specificity
As shown herein, CRISPR-Cas RNA-guided nucleases based on the S.
pyogenes Cas9 protein can have significant off-target mutagenic effects that are comparable to or higher than the intended on-target activity (Example 1). Such off- target effects can be problematic for research and in particular for potential therapeutic applications. Therefore, methods for improving the specificity of
CRISPR-Cas RNA guided nucleases (RGNs) are needed.
As described in Example 1 , Cas9 RGNs can induce high-frequency indel mutations at off-target sites in human cells (see also Cradick et al., 2013; Fu et al., 2013; Hsu et al., 2013; Pattanayak et al, 2013). These undesired alterations can occur at genomic sequences that differ by as many as five mismatches from the intended on- target site (see Example 1). In addition, although mismatches at the 5' end of the gRNA complementarity region are generally better tolerated than those at the 3 ' end, these associations are not absolute and show site-to-site-dependence (see Example 1 and Fu et al., 2013; Hsu et al., 2013; Pattanayak et al., 2013). As a result,
computational methods that rely on the number and/or positions of mismatches currently have limited predictive value for identifying bona fide off-target sites.
Therefore, methods for reducing the frequencies of off-target mutations remain an important priority if RNA-guided nucleases are to be used for research and therapeutic applications.
Dimerization is an attractive potential strategy for improving the specificity of Cas9 nucleases. This is distinct from a paired Cas9 nickase approach, which is not a true dimeric system. Paired nickases work by co-localizing two Cas9 nickases on a segment of DNA, thereby inducing high efficiency genome editing via an undefined mechanism. Because dimerization is not required for enzymatic activity, single Cas9 nickases can also induce indels with high efficiencies at certain sites (via an unknown mechanism) and can therefore potentially cause unwanted off-target mutations in the genome.
Thus, one strategy to improve the specificity of RGNs is fusing a Fokl endonuclease domain to a catalytically inactive form of Cas9 bearing the D10A and H840A mutations (also known as dCas9). Fokl nuclease domain functions as a dimer and therefore two subunits must be recruited to the same local piece of DNA in order to induce a double-stranded break. In this configuration (Figure 9A and Example 2), two FokI-dCas9 fusions are recruited in an appropriate configuration using two different gRNAs to yield a double-stranded break. Thus, in this system, the Fokl- dCas9 fusions would bind to a site that is twice as long as that of a single RGN and therefore this system would be expected to be more specific.
Therefore provided herein are FokI-dCas9 fusion proteins, wherein the Fokl sequence is fused to dCas9 (preferably to the amino-terminal end of dCas9, but also optionally to the carboxy terminus), optionally with an intervening linker, e.g., a linker of from 2-30 amino acids, e.g., 4-12 amino acids, e.g., Gly4Ser (SEQ ID NO:23) or (Gly4Ser)3. In some embodiments, the fusion proteins include a linker between the dCas9 and the Fokl domains. Linkers that can be used in these fusion proteins (or between fusion proteins in a concatenated structure) can include any sequence that does not interfere with the function of the fusion proteins. In preferred embodiments, the linkers are short, e.g., 2-20 amino acids, and are typically flexible (i.e., comprising amino acids with a high degree of freedom such as glycine, alanine, and serine). In some embodiments, the linker comprises one or more units consisting of GGGS (SEQ ID NO:22) or GGGGS (SEQ ID NO:23), e.g., two, three, four, or more repeats of the GGGS (SEQ ID NO:22) or GGGGS (SEQ ID NO:23) unit. Other linker sequences can also be used. Also described herein is a RNA-guided Fokl nuclease platform in which dimerization, rather than just co-localization, is required for efficient genome editing activity. These nucleases can robustly mediate highly efficient genome editing in human cells and can reduce off-target mutations to undetectable levels as judged by sensitive deep sequencing methods. Also described is an efficient system for expressing pairs of gRNAs with any 5 ' end nucleotide, a method that confers a wider targeting range on the RFN platform. Finally, monomeric Cas9 nickases generally introduce more undesirable indels and point mutations than the nucleases described herein in the presence of a single gRNA. These results define a robust, user-friendly nuclease platform with the specificity advantages of a well-characterized dimeric architecture and an improved mutagenesis profile relative to paired Cas9 nickases, features that will be important for research or therapeutic applications requiring the highest possible genome editing precision.
Thus a new RNA-guided Fokl Nuclease (RFN) platform is described herein for performing robust and highly specific genome editing in human cells. RFNs require two gRNAs for activity and function as dimers. Surprisingly, the engineering of an active RFN required fusion of the Fokl nuclease domain to the amino-terminal end of the dCas9 protein, an architecture different from ZFNs and TALENs in which the Fokl domain is fused to the carboxy-terminal end of engineered zinc finger or transcription activator-like effector repeat arrays. RFNs also require that the half-sites bound by each Fok-dCas9/gRNA complex have a particular relative orientation (PAMs out) with a relatively restricted intervening spacer length of 14 to 17 bps (although activity may be possible at additional spacings but with less consistent success).
The dimeric nature of RFNs provides important specificity advantages relative to standard monomeric Cas9 nucleases. In an ideal dimeric system, little to no activity will be observed with monomers on half-sites. The present data demonstrate that FokI-dCas9 directed by a single gRNA induces very little or no mutagenesis at RFN half-sites. 12 single gRNAs (for six RFN target sites) were tested with co- expressed FokI-dCas9 and indels were observed at very low frequencies (range of 0.0045% to 0.47%), in some cases at levels as low as background rates observed in control cells in which there was no expression of gRNA or nuclease. Given that the Fokl nuclease domain functions as a dimer, it is presumed that any indels observed with a single gR A are likely due to recruitment of a FokI-dCas9 dimer to the DNA. Regardless of mechanism, given that only very low level mutagenesis was observed when FokI-dCas9 was tested with single gRNAs at 12 on-target half-sites, it is very unlikely that any mutagenesis will be induced at partially mismatched, off-target half- sites. Indeed, an RFN targeted to VEGFA did not induce detectable mutations at known off-target sites of one of the gRNAs as judged by deep sequencing.
Because RFNs are a true dimeric system, they possess a number of important advantages over paired nickase technology, which depends on co-localization but does not require dimerization. First, the direct comparisons herein show that single Cas9 nickases generally induce indel mutations with greater efficiencies than do Fokl- dCas9 fusion proteins directed by the same individual gRNAs. Second, monomeric Cas9 nickases can also induce base pair substitutions in their target half-sites with high efficiencies, a previously unknown mutagenic side-effect that we uncovered in this study. Again, the direct comparisons show that monomeric Cas9 nickases induce these unwanted point mutations at substantially higher rates than FokI-dCas9 fusions guided by the same single gRNAs. Third, paired Cas9 nickases show greater promiscuity in the orientation and spacing of target half-sites than dimeric RFNs and therefore have a greater potential range of sites at which off-target mutations might be induced. Paired nickase half-sites can be oriented with their PAMs in or PAMs out and with spacer sequences ranging in length from 0 to 1000 bps (Ran et al, Cell 154, 1380-1389 (2013); Mali et al, Nat Biotechnol 31, 833-838 (2013); Cho et al, Genome Res (2013)). This promiscuity exists because the genome editing activities of Cas9 nickases do not depend on dimerization of the enzyme but rather just co- localization of the two nicks. By contrast, RFNs are much more stringent in their specificities— half-sites must have their PAMs out and must be spaced apart by 14 to 17 bps, due to the requirement for two appropriately positioned Fokl cleavage domains for efficient cleavage.
Fokl
Fokl is a type lis restriction endonuclease that includes a DNA recognition domain and a catalytic (endonuclease) domain. The fusion proteins described herein can include all of Fokl or just the catalytic endonuclease domain, e.g., amino acids 388-583 or 408-583 of GenBank Acc. No. AAA24927.1, e.g., as described in Li et al, Nucleic Acids Res. 39(1): 359-372 (2011); Cathomen and Joung, Mol. Ther. 16: 1200-1207 (2008), or a mutated form of Fokl as described in Miller et al. Nat
Biotechnol 25: 778-785 (2007); Szczepek et al, Nat Biotechnol 25: 786-793 (2007); or Bitinaite et al, Proc. Natl. Acad. Sci. USA. 95: 10570-10575 (1998).
An exemplary amino acid sequence of Fokl is as follows:
10 20 30 40 50 60
MFLSMVSKIR TFGWVQNPGK FENLKRWQV FDRNSKVHNE VKNIKIPTLV KESKIQKELV
70 80 90 100 110 120
AIMNQHDLIY TYKELVGTGT SIRSEAPCDA I IQATIADQG NKKGYIDNWS SDGFLRWAHA
130 140 150 160 170 180
LGFIEYINKS DSFVITDVGL AYSKSADGSA IEKEILIEAI SSYPPAIRIL TLLEDGQHLT
190 200 210 220 230 240
KFDLGKNLGF SGESGFTSLP EGILLDTLAN AMPKDKGEIR NNWEGSSDKY ARMIGGWLDK
250 260 270 280 290 300
LGLVKQGKKE FIIPTLGKPD NKEFISHAFK ITGEGLKVLR RAKGSTKFTR VPKRVYWEML
310 320 330 340 350 360
ATNLTDKEYV RTRRALILEI LIKAGSLKIE QIQDNLKKLG FDEVIETIEN DIKGLINTGI
370 380 390 400 410 420
FIEIKGRFYQ LKDHILQFVI PNRGVTKQLV KSELEEKKSE LRHKLKYVPH EYIELIEIAR
430 440 450 460 470 480
NSTQDRILEM KVMEFFMKVY GYRGKHLGGS RKPDGAIYTV GSPIDYGVIV DTKAYSGGYN
490 500 510 520 530 540
LPIGQADEMQ RYVEENQTRN KHINPNEWWK VYPSSVTEFK FLFVSGHFKG NYKAQLTRLN
550 560 570 580
HITNCNGAVL SVEELLIGGE MIKAGTLTLE EVRRKFNNGE INF (SEQ ID NO : 4 )
An exemplary nucleic acid sequence encoding Fokl is as follows:
ATGTTTTTGAGTATGGTTTCTAAAATAAGAACTTTCGGTTGGGTTCAAAATCCAGGTAAA TTTGAGAATTTAAAACGAGTAGTTCAAGTATTTGATAGAAATTCTAAAGTACATAATGAA GTGAAAAATATAAAGATACCAACCCTAGTCAAAGAAAGTAAGATCCAAAAAGAACTAGTT GCTATTATGAATCAACATGATTTGATTTATACATATAAAGAGTTAGTAGGAACAGGAACT TCAATACGTTCAGAAGCACCATGCGATGCAATTATTCAAGCAACAATAGCAGATCAAGGA AATAAAAAAGGCTATATCGATAATTGGTCATCTGACGGTTTTTTGCGTTGGGCACATGCT TTAGGATTTATTGAATATATAAATAAAAGTGATTCTTTTGTAATAACTGATGTTGGACTT GCTTACTCTAAATCAGCTGACGGCAGCGCCATTGAAAAAGAGATTTTGATTGAAGCGATA TCATCTTATCCTCCAGCGATTCGTATTTTAACTTTGCTAGAAGATGGACAACATTTGACA AAGTTTGATCTTGGCAAGAATTTAGGTTTTAGTGGAGAAAGTGGATTTACTTCTCTACCG GAAGGAATTCTTTTAGATACTCTAGCTAATGCTATGCCTAAAGATAAAGGCGAAATTCGT AATAATTGGGAAGGATCTTCAGATAAGTACGCAAGAATGATAGGTGGTTGGCTGGATAAA CTAGGATTAGTAAAGCAAGGAAAAAAAGAATTTATCATTCCTACTTTGGGTAAGCCGGAC AATAAAGAGTTTATATCCCACGCTTTTAAAATTACTGGAGAAGGTTTGAAAGTACTGCGT CGAGCAAAAGGCTCTACAAAATTTACACGTGTACCTAAAAGAGTATATTGGGAAATGCTT GCTACAAACCTAACCGATAAAGAGTATGTAAGAACAAGAAGAGCTTTGATTTTAGAAATA TTAATCAAAGCTGGATCATTAAAAATAGAACAAATACAAGACAACTTGAAGAAATTAGGA TTTGATGAAGTTATAGAAACTATTGAAAATGATATCAAAGGCTTAATTAACACAGGTATA TTTATAGAAATCAAAGGGCGATTTTATCAATTGAAAGACCATATTCTTCAATTTGTAATA CCTAATCGTGGTGTGACTAAGCAACTAGTCAAAAGTGAACTGGAGGAGAAGAAATCTGAA CTTCGTCATAAATTGAAATATGTGCCTCATGAATATATTGAATTAATTGAAATTGCCAGA AATTCCACTCAGGATAGAATTCTTGAAATGAAGGTAATGGAATTTTTTATGAAAGTTTAT
GGATATAGAGGTAAACATTTGGGTGGATCAAGGAAACCGGACGGAGCAATTTATACTGTC GGATCTCCTATTGATTACGGTGTGATCGTGGATACTAAAGCTTATAGCGGAGGTTATAAT CTGCCAATTGGCCAAGCAGATGAAATGCAACGATATGTCGAAGAAAATCAAACACGAAAC AAACATATCAACCCTAATGAATGGTGGAAAGTCTATCCATCTTCTGTAACGGAATTTAAG TTTTTATTTGTGAGTGGTCAC TTAAAGGAAACTACAAAGCTCAGCTTACACGATTAAAT CATATCACTAATTGTAATGGAGCTGTTCTTAGTGTAGAAGAGCTTTTAATTGGTGGAGAA ATGATTAAAGCCGGCACATTAACCTTAGAGGAAGTGAGACGGAAATTTAATAACGGCGAG A AAAC AA (SEQ ID NO: 285)
In some embodiments, the Fokl nuclease used herein is at least about 50% identical SEQ ID NO:4, e.g., to amino acids 388-583 or 408-583 of SEQ ID NO:4. These variant nucleases must retain the ability to cleave DNA. In some embodiments, the nucleotide sequences are about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identical to amino acids 388-583 or 408-583 of SEQ ID NO:4. In some embodiments, any differences from amino acids 388-583 or 408-583 of SEQ ID NO:4 are in non-conserved regions.
To determine the percent identity of two sequences, the sequences are aligned for optimal comparison purposes (gaps are introduced in one or both of a first and a second amino acid or nucleic acid sequence as required for optimal alignment, and non-homologous sequences can be disregarded for comparison purposes). The length of a reference sequence aligned for comparison purposes is at least 50%> (in some embodiments, about 50%, 55%, 60%, 65%, 70%, 75%, 85%, 90%, 95%, or 100% of the length of the reference sequence is aligned). The nucleotides or residues at corresponding positions are then compared. When a position in the first sequence is occupied by the same nucleotide or residue as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For purposes of the present application, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J. Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package, using a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5. Cas9
A number of bacteria express Cas9 protein variants. The Cas9 from
Streptococcus pyogenes is presently the most commonly used; some of the other Cas9 proteins have high levels of sequence identity with the S. pyogenes Cas9 and use the same guide R As. Others are more diverse, use different gR As, and recognize different PAM sequences as well (the 2-5 nucleotide sequence specified by the protein which is adjacent to the sequence specified by the RNA). Chylinski et al. classified Cas9 proteins from a large group of bacteria (RNA Biology 10:5, 1-12; 2013), and a large number of Cas9 proteins are listed in supplementary figure 1 and supplementary table 1 thereof, which are incorporated by reference herein. Additional Cas9 proteins are described in Esvelt et al., Nat Methods. 2013 Nov; 10(11): 1116-21 and Fonfara et al., "Phylogeny of Cas9 determines functional exchangeability of dual-RNA and Cas9 among orthologous type II CRISPR-Cas systems." Nucleic Acids Res. 2013 Nov 22. [Epub ahead of print] doi: 10.1093/nar/gktl074.
Cas9 molecules of a variety of species can be used in the methods and compositions described herein. While the S. pyogenes and S. thermophilus Cas9 molecules are the subject of much of the disclosure herein, Cas9 molecules of, derived from, or based on the Cas9 proteins of other species listed herein can be used as well. In other words, while the much of the description herein uses S. pyogenes and S. thermophilus Cas9 molecules, Cas9 molecules from the other species can replace them. Such species include those set forth in the following table, which was created based on supplementary figure 1 of Chylinski et al, 2013.
Figure imgf000021_0001
Alternative Cas9 proteins
GenBank Acc No. Bacterium
302336020 Olsenella uli DSM 7084
366983953 Oenococcus kitaharae DSM 17330
310286728 Bifidobacterium bifidum SI 7
258509199 Lactobacillus rhamnosus GG
300361537 Lactobacillus gasseri JV-V03
169823755 Finegoldia magna ATCC 29328
47458868 Mycoplasma mobile 163K
284931710 Mycoplasma gallisepticum str. F
363542550 Mycoplasma ovipneumoniae SCOl
384393286 Mycoplasma canis PG 14
71894592 Mycoplasma synoviae 53
238924075 Eubacterium rectale ATCC 33656
116627542 Streptococcus thermophilus LMD-9
315149830 Enterococcus faecalis TX0012
315659848 Staphylococcus lugdunensis M23590
160915782 Eubacterium dolichum DSM 3991
336393381 Lactobacillus coryniformis subsp. torquens
310780384 Ilyobacter polytropus DSM 2926
325677756 Ruminococcus albus 8
187736489 Akkermansia muciniphila ATCC BAA-835
117929158 Acidothermus cellulolyticus 11B
189440764 Bifidobacterium longum D JO 10 A
283456135 Bifidobacterium dentium Bdl
38232678 Corynebacterium diphtheriae NCTC 13129
187250660 Elusimicrobium minutum Peil91
319957206 Nitratifractor salsuginis DSM 16511
325972003 Sphaerochaeta globus str. Buddy
261414553 Fibrobacter succinogenes subsp. succinogenes
60683389 Bacteroides fragilis NCTC 9343
256819408 Capnocytophaga ochracea DSM 7271
90425961 Rhodopseudomonas palustris BisB18
373501184 Prevotella micans F0438
294674019 Prevotella ruminicola 23
365959402 Flavobacterium columnare ATCC 49512
312879015 Aminomonas paucivorans DSM 12260
83591793 Rhodospirillum rubrum ATCC 11170
294086111 Candidatus Puniceispirillum marinum IMCC1322
121608211 Verminephrobacter eiseniae EF01-2
344171927 Ralstonia syzygii R24
159042956 Dinoroseobacter shibae DFL 12
288957741 Azospirillum sp- B510
92109262 Nitrobacter hamburgensis XI 4
148255343 Bradyrhizobium sp- BTAil
34557790 Wolinella succinogenes DSM 1740
218563121 Campylobacter jejuni subsp. jejuni
291276265 Helicobacter mustelae 12198 Alternative Cas9 proteins
GenBank Acc No. Bacterium
229113166 Bacillus cereus Rockl-15
222109285 Acidovorax ebreus TPSY
189485225 uncultured Termite group 1
182624245 Clostridium perfringens D str.
220930482 Clostridium cellulolyticum H10
154250555 Parvibaculum lavamentivorans DS-1
257413184 Roseburia intestinalis LI -82
218767588 Neisseria meningitidis Z2491
15602992 Pasteurella multocida subsp. multocida
319941583 Sutterella wadsworthensis 3 1
254447899 gamma proteobacterium HTCC5015
54296138 Legionella pneumophila str. Paris
331001027 Parasutterella excrementihominis YIT 11859
34557932 Wolinella succinogenes DSM 1740
118497352 Francisella novicida U112
The constructs and methods described herein can include the use of any of those Cas9 proteins, and their corresponding guide RNAs or other guide RNAs that are compatible. The Cas9 from Streptococcus thermophilus LMD-9 CRISPR1 system has also been shown to function in human cells in Cong et al (Science 339, 819 (2013)). Cas9 orthologs from N. meningitides are described in Hou et al, Proc Natl Acad Sci U S A. 2013 Sep 24;110(39): 15644-9 and Esvelt et al, Nat Methods. 2013 Nov;10(l 1): 1116-21. Additionally, Jinek et al. showed in vitro that Cas9 orthologs from S. thermophilus and L. innocua, (but not from N. meningitidis or C. jejuni, which likely use a different guide RNA), can be guided by a dual S. pyogenes gRNA to cleave target plasmid DNA, albeit with slightly decreased efficiency.
In some embodiments, the present system utilizes the Cas9 protein from S. pyogenes, either as encoded in bacteria or codon-optimized for expression in mammalian cells, containing mutations at D10, E762, H983, or D986 and H840 or N863, e.g., D10A/D10N and H840A/H840N/H840Y, to render the nuclease portion of the protein catalytically inactive; substitutions at these positions could be alanine (as they are in Nishimasu al, Cell 156, 935-949 (2014)) or they could be other residues, e.g., glutamine, asparagine, tyrosine, serine, or aspartate, e.g.,, E762Q, H983N, H983Y, D986N, N863D, N863S, or N863H (Figure 1C). The sequence of the catalytically inactive S. pyogenes Cas9 that can be used in the methods and compositions described herein is as follows; the exemplary mutations of D10A and H840A are in bold and underlined. 10 20 30 40 50 60
MDKKYSIGLA IGTNSVGWAV ITDEYKVPSK KFKVLGNTDR HSIKKNLIGA LLFDSGETAE
70 80 90 100 110 120
ATRLKRTARR RYTRRKNRIC YLQEIFSNEM AKVDDSFFHR LEESFLVEED KKHERHPIFG
130 140 150 160 170 180
NIVDEVAYHE KYPTIYHLRK KLVDSTDKAD LRLIYLALAH MIKFRGHFLI EGDLNPDNSD
190 200 210 220 230 240
VDKLFIQLVQ TYNQLFEENP INASGVDAKA ILSARLSKSR RLENLIAQLP GEKKNGLFGN
250 260 270 280 290 300
LIALSLGLTP NFKSNFDLAE DAKLQLSKDT YDDDLDNLLA QIGDQYADLF LAAKNLSDAI
310 320 330 340 350 360
LLSDILRVNT EITKAPLSAS MIKRYDEHHQ DLTLLKALVR QQLPEKYKEI FFDQSKNGYA
370 380 390 400 410 420
GYIDGGASQE EFYKFIKPIL EKMDGTEELL VKLNREDLLR KQRTFDNGSI PHQIHLGELH
430 440 450 460 470 480
AILRRQEDFY PFLKDNREKI EKILTFRIPY YVGPLARGNS RFAWMTRKSE ETITPWNFEE
490 500 510 520 530 540
WDKGASAQS FIERMTNFDK NLPNEKVLPK HSLLYEYFTV YNELTKVKYV TEGMRKPAFL
550 560 570 580 590 600
SGEQKKAIVD LLFKTNRKVT VKQLKEDYFK KIECFDSVEI SGVEDRFNAS LGTYHDLLKI
610 620 630 640 650 660
IKDKDFLDNE ENEDILEDIV LTLTLFEDRE MIEERLKTYA HLFDDKVMKQ LKRRRYTGWG
670 680 690 700 710 720
RLSRKLINGI RDKQSGKTIL DFLKSDGFAN RNFMQLIHDD SLTFKEDIQK AQVSGQGDSL
730 740 750 760 770 780
HEHIANLAGS PAIKKGILQT VKWDELVKV MGRHKPENIV IEMARENQTT QKGQKNSRER
790 800 810 820 830 840
MKRIEEGIKE LGSQILKEHP VENTQLQNEK LYLYYLQNGR DMYVDQELDI NRLSDYDVDA
850 860 870 880 890 900
IVPQSFLKDD SIDNKVLTRS DKNRGKSDNV PSEEWKKMK NYWRQLLNAK LITQRKFDNL
910 920 930 940 950 960
TKAERGGLSE LDKAGFIKRQ LVETRQITKH VAQILDSRMN TKYDENDKLI REVKVITLKS
970 980 990 1000 1010 1020
KLVSDFRKDF QFYKVREINN YHHAHDAYLN AWGTALIKK YPKLESEFVY GDYKVYDVRK
1030 1040 1050 1060 1070 1080
MIAKSEQEIG KATAKYFFYS NIMNFFKTEI TLANGEIRKR PLIETNGETG EIVWDKGRDF
1090 1100 1110 1120 1130 1140
ATVRKVLSMP QVNIVKKTEV QTGGFSKESI LPKRNSDKLI ARKKDWDPKK YGGFDSPTVA
1150 1160 1170 1180 1190 1200
YSVLWAKVE KGKSKKLKSV KELLGITIME RSSFEKNPID FLEAKGYKEV KKDLI IKLPK
1210 1220 1230 1240 1250 1260 YSLFELENGR KRMLASAGEL QKGNELALPS KYVNFLYLAS HYEKLKGSPE DNEQKQLFVE
1270 1280 1290 1300 1310 1320
QHKHYLDEI I EQISEFSKRV ILADANLDKV LSAYNKHRDK PIREQAE I I HLFTLTNLGA
1330 1340 1350 1360
PAAFKYFDTT IDRKRYTSTK EVLDATLIHQ SITGLYETRI DLSQLGGD (SEQ ID NO: 5)
In some embodiments, the Cas9 nuclease used herein is at least about 50% identical to the sequence of S. pyogenes Cas9, i.e., at least 50% identical to SEQ ID NO:5. In some embodiments, the nucleotide sequences are about 50%>, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identical to SEQ ID NO:5. In some embodiments, any differences from SEQ ID NO:5 are in non-conserved regions, as identified by sequence alignment of sequences set forth in Chylinski et al, RNA Biology 10:5, 1-12; 2013 (e.g., in supplementary figure 1 and supplementary table 1 thereof); Esvelt et al, Nat Methods. 2013 Nov; 10(11): 1116-21 and Fonfara et al, Nucl. Acids Res. (2014) 42 (4): 2577-2590. [Epub ahead of print 2013 Nov 22] doi: 10.1093/nar/gktl074. Identity is determined as set forth above.
Guide RNAs (gRNAs)
Guide RNAs generally speaking come in two different systems: System 1, which uses separate crRNA and tracrRNAs that function together to guide cleavage by Cas9, and System 2, which uses a chimeric crRNA-tracrRNA hybrid that combines the two separate guide RNAs in a single system (referred to as a single guide RNA or sgRNA, see also Jinek et al., Science 2012; 337:816-821). The tracrRNA can be variably truncated and a range of lengths has been shown to function in both the separate system (system 1) and the chimeric gRNA system (system 2). For example, in some embodiments, tracrRNA may be truncated from its 3' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts. In some embodiments, the tracrRNA molecule may be truncated from its 5' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts. Alternatively, the tracrRNA molecule may be truncated from both the 5' and 3' end, e.g., by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 nts on the 5' end and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts on the 3' end. See, e.g., Jinek et al, Science 2012; 337:816-821; Mali et al, Science. 2013 Feb 15;339(6121):823-6; Cong et al, Science. 2013 Feb 15;339(6121):819-23; and Hwang and Fu et al, Nat Biotechnol. 2013 Mar;31(3):227-9; Jinek et al, Elife 2, e00471 (2013)). For System 2, generally the longer length chimeric gRNAs have shown greater on-target activity but the relative specificities of the various length gR As currently remain undefined and therefore it may be desirable in certain instances to use shorter gRNAs. In some embodiments, the gRNAs are
complementary to a region that is within about 100-800 bp upstream of the transcription start site, e.g., is within about 500 bp upstream of the transcription start site, includes the transcription start site, or within about 100-800 bp, e.g., within about 500 bp, downstream of the transcription start site. In some embodiments, vectors (e.g., plasmids) encoding more than one gRNA are used, e.g., plasmids encoding, 2, 3, 4, 5, or more gRNAs directed to different sites in the same region of the target gene.
Cas9 nuclease can be guided to specific 17-20 nt genomic targets bearing an additional proximal protospacer adjacent motif (PAM), e.g., of sequence NGG, using a guide RNA, e.g., a single gRNA or a tracrRNA/crRNA, bearing 17-20 nts at its 5' end that are complementary to the complementary strand of the genomic DNA target site. Thus, the present methods can include the use of a single guide RNA comprising a crRNA fused to a normally trans-encoded tracrRNA, e.g., a single Cas9 guide RNA as described in Mali et al, Science 2013 Feb 15; 339(6121):823-6, with a sequence at the 5' end that is complementary to the target sequence, e.g., of 25-17, optionally 20 or fewer nucleotides (nts), e.g., 20, 19, 18, or 17 nts, preferably 17 or 18 nts, of the complementary strand to a target sequence immediately 5 Of a protospacer adjacent motif (PAM), e.g., NGG, NAG, or NNGG In some embodiments, the single Cas9 guide RNA consists of the sequence:
(Xi7-2o)GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUC CG(XN) (SEQ ID NO:6);
(Xi7-2o)GUUUUAGAGCUAUGCUGAAAAGCAUAGCAAGUUAAAAUAAGGCU AGUCCGUUAUC(XN) (SEQ ID NO:7);
(Xi7-2o)GUUUUAGAGCUAUGCUGUUUUGGAAACAAAACAGCAUAGCAAGU UAAAAUAAGGCUAGUCCGUUAUC(XN) (SEQ ID NO: 8);
(Xi7-2o)GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUU AUCAACUUGAAAAAGUGGCACCGAGUCGGUGC(XN) (SEQ ID NO:9), (Xi7-2o)GUUUAAGAGCUAGAAAUAGCAAGUUUAAAUAAGGCUAGUCCGUU AUCAACUUGAAAAAGUGGCACCGAGUCGGUGC(SEQ ID NO: 10);
(Xi7-2o)GUUUUAGAGCUAUGCUGGAAACAGCAUAGCAAGUUUAAAUAAGG CUAGUCC GUUAUC A ACUUG AAAAAGUGGC AC CG AGUC GGUGC (SEQ ID NO: 11); or
(Xi7-2o)GUUUAAGAGCUAUGCUGGAAACAGCAUAGCAAGUUUAAAUAAGG CUAGUCC GUUAUC A ACUUG AAAAAGUGGC AC CG AGUC GGUGC (SEQ ID NO: 12);
wherein X17-20 is the nucleotide sequence complementary to 17-20 consecutive nucleotides of the target sequence. DNAs encoding the single guide RNAs have been described previously in the literature (Jinek et al, Science. 337(6096):816-21 (2012) and Jinek et al, Elife. 2:e00471 (2013)).
The guide RNAs can include XN which can be any sequence, wherein N (in the RNA) can be 0-200, e.g., 0-100, 0-50, or 0-20, that does not interfere with the binding of the ribonucleic acid to Cas9.
In some embodiments, the guide RNA includes one or more Adenine (A) or Uracil (U) nucleotides on the 3 ' end. In some embodiments the RNA includes one or more U, e.g., 1 to 8 or more Us (e.g., U, UU, UUU, UUUU, UUUUU, UUUUUU, UUUUUUU, UUUUUUUU) at the 3' end of the molecule, as a result of the optional presence of one or more Ts used as a termination signal to terminate RNA PolIII transcription.
Although some of the examples described herein utilize a single gRNA, the methods can also be used with dual gRNAs (e.g., the crRNA and tracrRNA found in naturally occurring systems). In this case, a single tracrRNA would be used in conjunction with multiple different crRNAs expressed using the present system, e.g., the following:
(Xi7_2o)GUUUUAGAGCUA (SEQ ID NO: 13);
(Xi7-2o)GUUUUAGAGCUAUGCUGUUUUG (SEQ ID NO: 14); or
(Xi7-2o)GUUUUAGAGCUAUGCU (SEQ ID NO: 15); and a tracrRNA sequence. In this case, the crRNA is used as the guide RNA in the methods and molecules described herein, and the tracrRNA can be expressed from the same or a different DNA molecule. In some embodiments, the methods include contacting the cell with a tracrRNA comprising or consisting of the sequence
GGAACCAUUCAAAACAGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUA UC A ACUUG AAAAAGUGGC AC C G AGUCGGUGC (SEQ ID NO: 16) or an active portion thereof (an active portion is one that retains the ability to form complexes with Cas9 or dCas9). In some embodiments, the tracrRNA molecule may be truncated from its 3' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts. In another embodiment, the tracrR A molecule may be truncated from its 5 ' end by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts. Alternatively, the tracrRNA molecule may be truncated from both the 5' and 3' end, e.g., by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 nts on the 5' end and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35 or 40 nts on the 3' end. Exemplary tracrRNA sequences in addition to SEQ ID NO: 8 include the following:
UAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCA
CCGAGUCGGUGC (SEQ ID NO: 17) or an active portion thereof; or
AGC AUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUC AACUUGAAAAAGU
GGCACCGAGUCGGUGC (SEQ ID NO: 18) or an active portion thereof.
In some embodiments wherein (Xi7-2o)GUUUUAGAGCUAUGCUGUUUUG
(SEQ ID NO: 14) is used as a crRNA, the following tracrRNA is used:
GGAACCAUUCAAAACAGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUA UC A ACUUG AAAA AGUGGC AC C G AGUCGGUGC (SEQ ID NO : 16) or an active portion thereof. In some embodiments wherein (Xi7_2o)GUUUUAGAGCUA (SEQ ID
NO: 13) is used as a crRNA, the following tracrRNA is used:
UAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCA CCGAGUCGGUGC (SEQ ID NO: 17) or an active portion thereof. In some embodiments wherein (Xi7_20)GUUUUAGAGCUAUGCU (SEQ ID NO : 15) is used as a crRNA, the following tracrRNA is used:
AGCAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGU GGCACCGAGUCGGUGC (SEQ ID NO: 18) or an active portion thereof.
In some embodiments, the gRNA is targeted to a site that is at least three or more mismatches different from any sequence in the rest of the genome in order to minimize off-target effects.
Modified RNA oligonucleotides such as locked nucleic acids (LNAs) have been demonstrated to increase the specificity of RNA-DNA hybridization by locking the modified oligonucleotides in a more favorable (stable) conformation. For example, 2'-0-methyl RNA is a modified base where there is an additional covalent linkage between the 2' oxygen and 4' carbon which when incorporated into oligonucleotides can improve overall thermal stability and selectivity (Formula I).
Figure imgf000029_0001
Formula I - Locked Nucleic Acid
Thus in some embodiments, the tru-gRNAs disclosed herein may comprise one or more modified RNA oligonucleotides. For example, the truncated guide RNAs molecules described herein can have one, some or all of the 17-18 or 17-19 nts 5 ' region of the guideRNA complementary to the target sequence are modified, e.g., locked (2'-0-4'-C methylene bridge), 5'-methylcytidine, 2'-0-methyl-pseudouridine, or in which the ribose phosphate backbone has been replaced by a polyamide chain (peptide nucleic acid), e.g., a synthetic ribonucleic acid.
In other embodiments, one, some or all of the nucleotides of the tru-gRNA sequence may be modified, e.g., locked (2'-0-4'-C methylene bridge), 5'- methylcytidine, 2'-0-methyl-pseudouridine, or in which the ribose phosphate backbone has been replaced by a polyamide chain (peptide nucleic acid), e.g., a synthetic ribonucleic acid.
In some embodiments, the single guide RNAs and/or crRNAs and/or tracrRNAs can include one or more Adenine (A) or Uracil (U) nucleotides on the 3 ' end.
Existing Cas9-based RGNs use gRNA-DNA heteroduplex formation to guide targeting to genomic sites of interest. However, RNA-DNA heteroduplexes can form a more promiscuous range of structures than their DNA-DNA counterparts. In effect, DNA-DNA duplexes are more sensitive to mismatches, suggesting that a DNA- guided nuclease may not bind as readily to off-target sequences, making them comparatively more specific than RNA-guided nucleases. Thus, the guide RNAs usable in the methods described herein can be hybrids, i.e., wherein one or more deoxyribonucleotides, e.g., a short DNA oligonucleotide, replaces all or part of the gRNA, e.g., all or part of the complementarity region of a gRNA. This DNA-based molecule could replace either all or part of the gRNA in a single gRNA system or alternatively might replace all of part of the crRNA and/or tracrRNA in a dual crRNA/tracrRNA system. Such a system that incorporates DNA into the
complementarity region should more reliably target the intended genomic DNA sequences due to the general intolerance of DNA-DNA duplexes to mismatching compared to R A-DNA duplexes. Methods for making such duplexes are known in the art, See, e.g., Barker et al, BMC Genomics. 2005 Apr 22;6:57; and Sugimoto et al, Biochemistry. 2000 Sep 19;39(37): 11270-81.
In addition, in a system that uses separate crR A and tracrR A, one or both can be synthetic and include one or more modified (e.g., locked) nucleotides or deoxyribonucleotides .
In a cellular context, complexes of Cas9 with these synthetic gR As could be used to improve the genome-wide specificity of the CRISPR/Cas9 nuclease system.
The methods described can include expressing in a cell, or contacting the cell with, a Cas9 gRNA plus a fusion protein as described herein.
Expression Systems
In order to use the fusion proteins described, it may be desirable to express them from a nucleic acid that encodes them. This can be performed in a variety of ways. For example, the nucleic acid encoding the guide RNA can be cloned into an intermediate vector for transformation into prokaryotic or eukaryotic cells for replication and/or expression. Intermediate vectors are typically prokaryote vectors, e.g., plasmids, or shuttle vectors, or insect vectors, for storage or manipulation of the nucleic acid encoding the fusion proteins for production of the fusion proteins. The nucleic acid encoding the fusion proteins can also be cloned into an expression vector, for administration to a plant cell, animal cell, preferably a mammalian cell or a human cell, fungal cell, bacterial cell, or protozoan cell.
To obtain expression, a sequence encoding a fusion protein is typically subcloned into an expression vector that contains a promoter to direct transcription. Suitable bacterial and eukaryotic promoters are well known in the art and described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual (3d ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al, eds., 2010). Bacterial expression systems for expressing the engineered protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al, 1983, Gene 22:229-235). Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available. The promoter used to direct expression of a nucleic acid depends on the particular application. For example, a strong constitutive promoter is typically used for expression and purification of fusion proteins. In contrast, when the guide RNA is to be administered in vivo for gene regulation, either a constitutive or an inducible promoter can be used, depending on the particular use of the guide RNA. In addition, a preferred promoter for administration of the guide RNA can be a weak promoter, such as HSV TK or a promoter having similar activity. The promoter can also include elements that are responsive to transactivation, e.g., hypoxia response elements, Gal4 response elements, lac repressor response element, and small molecule control systems such as tetracycline-regulated systems and the RU-486 system (see, e.g., Gossen & Bujard, 1992, Proc. Natl. Acad. Sci. USA, 89:5547; Oligino et al, 1998, Gene Ther., 5:491-496; Wang et al, 1997, Gene Ther., 4:432-441; Neering et al, 1996, Blood, 88: 1147-55; and Rendahl et al, 1998, Nat. BiotechnoL, 16:757-761).
In addition to the promoter, the expression vector typically contains a transcription unit or expression cassette that contains all the additional elements required for the expression of the nucleic acid in host cells, either prokaryotic or eukaryotic. Atypical expression cassette thus contains a promoter operably linked, e.g., to the nucleic acid sequence encoding the gRNA, and any signals required, e.g., for efficient polyadenylation of the transcript, transcriptional termination, ribosome binding sites, or translation termination. Additional elements of the cassette may include, e.g., enhancers, and heterologous spliced intronic signals.
The particular expression vector used to transport the genetic information into the cell is selected with regard to the intended use of the gRNA, e.g., expression in plants, animals, bacteria, fungus, protozoa, etc. Standard bacterial expression vectors include plasmids such as pBR322 based plasmids, pSKF, pET23D, and commercially available tag-fusion expression systems such as GST and LacZ.
Expression vectors containing regulatory elements from eukaryotic viruses are often used in eukaryotic expression vectors, e.g., SV40 vectors, papilloma virus vectors, and vectors derived from Epstein-Barr virus. Other exemplary eukaryotic vectors include pMSG, pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 late promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.
The vectors for expressing the guide RNAs can include RNA Pol III promoters to drive expression of the guide RNAs, e.g., the HI, U6 or 7SK promoters. These human promoters allow for expression of gRNAs in mammalian cells following plasmid transfection. Alternatively, a T7 promoter may be used, e.g., for in vitro transcription, and the RNA can be transcribed in vitro and purified. Vectors suitable for the expression of short RNAs, e.g., siRNAs, shRNAs, or other small RNAs, can be used. With the Cys4-based multiplex system described in Figure 4B, multiple gRNAs can be expressed in a single transcript (driven by a RNA Pol II or Pol III promoter) and then cleaved out from that larger transcript.
Some expression systems have markers for selection of stably transfected cell lines such as thymidine kinase, hygromycin B phosphotransferase, and dihydrofolate reductase. High yield expression systems are also suitable, such as using a baculovirus vector in insect cells, with the gRNA encoding sequence under the direction of the polyhedrin promoter or other strong baculovirus promoters.
The elements that are typically included in expression vectors also include a replicon that functions in E. coli, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of recombinant sequences.
Standard transfection methods are used to produce bacterial, mammalian, yeast or insect cell lines that express large quantities of protein, which are then purified using standard techniques (see, e.g., Colley et al, 1989, J. Biol. Chem., 264: 17619-22; Guide to Protein Purification, in Methods in Enzymology, vol. 182 (Deutscher, ed., 1990)). Transformation of eukaryotic and prokaryotic cells are performed according to standard techniques (see, e.g., Morrison, 1977, J. Bacteriol. 132:349-351; Clark-Curtiss & Curtiss, Methods in Enzymology 101 :347-362 (Wu et al, eds, 1983).
Any of the known procedures for introducing foreign nucleotide sequences into host cells may be used. These include the use of calcium phosphate transfection, polybrene, protoplast fusion, electroporation, nucleofection, liposomes,
microinjection, naked DNA, plasmid vectors, viral vectors, both episomal and integrative, and any of the other well-known methods for introducing cloned genomic DNA, cDNA, synthetic DNA or other foreign genetic material into a host cell (see, e.g., Sambrook et al., supra). It is only necessary that the particular genetic engineering procedure used be capable of successfully introducing at least one gene into the host cell capable of expressing the gRNA.
The present invention includes the vectors and cells comprising the vectors.
EXAMPLES
The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.
Example 1. Assessing specificity of RNA-guided endonucleases
CRISPR RNA-guided nucleases (RGNs) have rapidly emerged as a facile and efficient platform for genome editing. This example describes the use of a human cell-based reporter assay to characterize off-target cleavage of Cas9-based RGNs. Materials and Methods
The following materials and methods were used in Example 1.
Construction of guide RNAs
DNA oligonucleotides harboring variable 20 nt sequences for Cas9 targeting were annealed to generate short double-strand DNA fragments with 4 bp overhangs compatible with ligation into BsmBI-digested plasmid pMLM3636. Cloning of these annealed oligonucleotides generates plasmids encoding a chimeric +103 single-chain guide RNA with 20 variable 5 ' nucleotides under expression of a U6 promoter
(Hwang et al, Nat Biotechnol 31 , 227-229 (2013); Mali et al, Science 339, 823-826 (2013)). pMLM3636 and the expression plasmid pJDS246 (encoding a codon optimized version of Cas9) used in this study are both available through the non-profit plasmid distribution service Addgene (addgene.org/crispr-cas).
EGFP Activity Assays
U20S.EGFP cells harboring a single integrated copy of an EGFP-PEST fusion gene were cultured as previously described (Reyon et al, Nat Biotech 30, 460- 465 (2012)). For transfections, 200,000 cells were Nucleofected with the indicated amounts of gRNA expression plasmid and pJDS246 together with 30 ng of a Td- tomato-encoding plasmid using the SE Cell Line 4D-Nucleofector™ X Kit (Lonza) according to the manufacturer's protocol. Cells were analyzed 2 days post- transfection using a BD LSRII flow cytometer. Transfections for optimizing gR A/Cas9 plasmid concentration were performed in triplicate and all other transfections were performed in duplicate.
PCR amplification and sequence verification of endogenous human genomic sites
PCR reactions were performed using Phusion Hot Start II high-fidelity DNA polymerase (NEB). Most loci amplified successfully using touchdown PCR (98 °C, 10 s; 72-62 °C, -1 °C/cycle, 15 s; 72 °C, 30 s]10 cycles, [98 °C, 10 s; 62 °C, 15 s; 72 °C, 30 s]25 cycles). PCR for the remaining targets were performed with 35 cycles at a constant annealing temperature of 68 °C or 72 °C and 3% DMSO or 1M betaine, if necessary. PCR products were analyzed on a QIAXCEL capillary electrophoresis system to verify both size and purity. Validated products were treated with ExoSap-IT (Affymetrix) and sequenced by the Sanger method (MGH DNA Sequencing Core) to verify each target site.
Determination of RGN-induced on- and off-target mutation frequencies in human cells
For U20S.EGFP and K562 cells, 2 x 105 cells were transfected with 250 ng of gRNA expression plasmid or an empty U6 promoter plasmid (for negative controls), 750 ng of Cas9 expression plasmid, and 30 ng of td-Tomato expression plasmid using the 4D Nucleofector System according to the manufacturer's instructions (Lonza). For HEK293 cells, 1.65 x 105 cells were transfected with 125 ng of gRNA expression plasmid or an empty U6 promoter plasmid (for the negative control), 375 ng of Cas9 expression plasmid, and 30 ng of a td-Tomato expression plasmid using
Lipofectamine LTX reagent according to the manufacturer's instructions (Life Technologies). Genomic DNA was harvested from transfected U20S.EGFP,
HEK293, or K562 cells using the QIAamp DNA Blood Mini Kit (QIAGEN), according to the manufacturer's instructions. To generate enough genomic DNA to amplify the off-target candidate sites, DNA from three Nucleofections (for
U20S.EGFP cells), two Nucleofections (for K562 cells), or two Lipofectamine LTX transfections was pooled together before performing T7EI. This was done twice for each condition tested, thereby generating duplicate pools of genomic DNA
representing a total of four or six individual transfections. PCR was then performed using these genomic DNAs as templates as described above and purified using Ampure XP beads (Agencourt) according to the manufacturer's instructions. T7EI assays were performed as previously described (Reyon et al, 2012, supra).
DNA sequencing of NHE J-mediated indel mutations
Purified PCR products used for the T7EI assay were cloned into Zero Blunt TOPO vector (Life Technologies) and plasmid DNAs were isolated using an alkaline lysis miniprep method by the MGH DNA Automation Core. Plasmids were sequenced using an Ml 3 forward primer (5' - GTAAAACGACGGCCAG - 3' (SEQ ID
NO: 19)) by the Sanger method (MGH DNA Sequencing Core).
Example la. Single Nucleotide Mismatches
To begin to define the specificity determinants of RGNs in human cells, a large-scale test was performed to assess the effects of systematically mismatching various positions within multiple gRNA/target DNA interfaces. To do this, a quantitative human cell-based enhanced green fluorescent protein (EGFP) disruption assay previously described (see Methods above and Reyon et al, 2012, supra) that enables rapid quantitation of targeted nuclease activities (Fig. 2B) was used. In this assay, the activities of nucleases targeted to a single integrated EGFP reporter gene can be quantified by assessing loss of fiuorescence signal in human U20S.EGFP cells caused by inactivating frameshift insertion/deletion (indel) mutations introduced by error prone non-homologous end-joining (NHEJ) repair of nuclease-induced double- stranded breaks (DSBs) (Fig. 2B). For the studies described here, three -100 nt single gRNAs (sgRNAs) targeted to different sequences within EGFP were used, as follows:
EGFP Site 1 GGGCACGGGCAGCTTGCCGGTGG (SEQ ID NO : 1 ) EGFP Site 2 GATGCCGTTCTTCTGCTTGTCGG (SEQ ID NO:2)
EGFP Site 3 GGTGGTGCAGATGAACTTCAGGG (SEQ ID NO:3)
Each of these sgRNAs can efficiently direct Cas9-mediated disruption of EGFP expression (see Example le and 2a, and FIGs. 3E (top) and 3F (top)).
In initial experiments, the effects of single nucleotide mismatches at 19 of 20 nucleotides in the complementary targeting region of three EG -targeted sgRNAs were tested. To do this, variant sgRNAs were generated for each of the three target sites harboring Watson-Crick transversion mismatches at positions 1 through 19 (numbered 1 to 20 in the 3' to 5' direction; see Fig. 1) and the abilities of these various sgR As to direct Cas9-mediated EGFP disruption in human cells tested (variant sgRNAs bearing a substitution at position 20 were not generated because this nucleotide is part of the U6 promoter sequence and therefore must remain a guanine to avoid affecting expression.)
For EGFP target site #2, single mismatches in positions 1 - 10 of the gRNA have dramatic effects on associated Cas9 activity (Fig. 2C, middle panel), consistent with previous studies that suggest mismatches at the 5 ' end of gRNAs are better tolerated than those at the 3' end (Jiang et al., Nat Biotechnol 31, 233-239 (2013); Cong et al, Science 339, 819-823 (2013); Jinek et al, Science 337, 816-821 (2012)). However, with EGFP target sites #1 and #3, single mismatches at all but a few positions in the gRNA appear to be well tolerated, even within the 3 ' end of the sequence. Furthermore, the specific positions that were sensitive to mismatch differed for these two targets (Fig. 2C, compare top and bottom panels) - for example, target site #1 was particularly sensitive to a mismatch at position 2 whereas target site #3 was most sensitive to mismatches at positions 1 and 8.
Example lb. Multiple Mismatches
To test the effects of more than one mismatch at the gRNA/DNA interface, a series of variant sgRNAs bearing double Watson-Crick transversion mismatches in adjacent and separated positions were created and the abilities of these sgRNAs to direct Cas9 nuclease activity were tested in human cells using the EGFP disruption assay. All three target sites generally showed greater sensitivity to double alterations in which one or both mismatches occur within the 3 ' half of the gRNA targeting region. However, the magnitude of these effects exhibited site-specific variation, with target site #2 showing the greatest sensitivity to these double mismatches and target site #1 generally showing the least. To test the number of adjacent mismatches that can be tolerated, variant sgRNAs were constructed bearing increasing numbers of mismatched positions ranging from positions 19 to 15 in the 5' end of the gRNA targeting region (where single and double mismatches appeared to be better tolerated).
Testing of these increasingly mismatched sgRNAs revealed that for all three target sites, the introduction of three or more adjacent mismatches results in significant loss of RGN activity. A sudden drop off in activity occurred for three different EGFP-targeted gRNAs as one makes progressive mismatches starting from position 19 in the 5' end and adding more mismatches moving toward the 3' end. Specifically, gRNAs containing mismatches at positions 19 and 19+18 show essentially full activity whereas those with mismatches at positions 19+18+17, 19+18+17+16, and 19+18+17+16+15 show essentially no difference relative to a negative control (Figure 2F). (Note that we did not mismatch position 20 in these variant gRNAs because this position needs to remain as a G because it is part of the U6 promoter that drives expression of the gRNA.)
Additional proof of that shortening gRNA complementarity might lead to RGNs with greater specificities was obtained in the following experiment: for four different EGFP-targeted gRNAs (Figure 2H), introduction of a double mismatch at positions 18 and 19 did not significantly impact activity. However, introduction of another double mismatch at positions 10 and 11 then into these gRNAs results in near complete loss of activity. Interestingly introduction of only the 10/11 double mismatches does not generally have as great an impact on activity.
Taken together, these results in human cells confirm that the activities of RGNs can be more sensitive to mismatches in the 3' half of the gRNA targeting sequence. However, the data also clearly reveal that the specificity of RGNs is complex and target site-dependent, with single and double mismatches often well tolerated even when one or more mismatches occur in the 3 ' half of the gRNA targeting region. Furthermore, these data also suggest that not all mismatches in the 5 ' half of the gRNA/DNA interface are necessarily well tolerated.
In addition, these results strongly suggest that gRNAs bearing shorter regions of complementarity (specifically ~17 nts) will be more specific in their activities. We note that 17 nts of specificity combined with the 2 nts of specificity conferred by the PAM sequence results in specification of a 19 bp sequence, one of sufficient length to be unique in large complex genomes such as those found in human cells.
Example lc. Off-Target Mutations
To determine whether off-target mutations for RGNs targeted to endogenous human genes could be identified, six sgRNAs that target three different sites in the VEGFA gene, one in the EMX1 gene, one in the RNF2 gene, and one in the FANCF gene were used. These six sgRNAs efficiently directed Cas9-mediated indels at their respective endogenous loci in human U20S.EGFP cells as detected by T7
Endonuclease I (T7EI) assay (Methods above). For each of these six RGNs, we then examined dozens of potential off-target sites (ranging in number from 46 to as many as 64) for evidence of nuclease-induced NHEJ-mediated indel mutations in
U20S.EGFP cells. The loci assessed included all genomic sites that differ by one or two nucleotides as well as subsets of genomic sites that differ by three to six nucleotides and with a bias toward those that had one or more of these mismatches in the 5' half of the gRNA targeting sequence. Using the T7EI assay, four off-target sites (out of 53 candidate sites examined) for VEGFA site 1, twelve (out of 46 examined) for VEGFA site 2, seven (out of 64 examined) for VEGFA site 3 and one (out of 46 examined) for the EMX1 site were readily identified. No off-target mutations were detected among the 43 and 50 potential sites examined for the RNF2 or FANCF genes, respectively. The rates of mutation at verified off-target sites were very high, ranging from 5.6% to 125% (mean of 40%) of the rate observed at the intended target site. These bona fide off-targets included sequences with mismatches in the 3' end of the target site and with as many as a total of five mismatches, with most off-target sites occurring within protein coding genes. DNA sequencing of a subset of off-target sites provided additional molecular confirmation that indel mutations occur at the expected RGN cleavage site (Figs. 8A-C).
Example Id. Off-Target Mutations in Other Cell Types
Having established that RGNs can induce off-target mutations with high frequencies in U20S.EGFP cells, it was next sought to determine whether these nucleases would also have these effects in other types of human cells. U20S.EGFP cells had been chosen for initial experiments because these cells were previously used to evaluate the activities of TALENs15 but human HEK293 and K562 cells have been more widely used to test the activities of targeted nucleases. Therefore, the activities of the four RGNs targeted to VEGFA sites 1, 2, and 3 and the EMX1 site were also assessed in HEK293 and K562 cells. Each of these four RGNs efficiently induced
NHEJ-mediated indel mutations at their intended on-target site in these two additional human cell lines (as assessed by T7EI assay), albeit with somewhat lower mutation frequencies than those observed in U20S.EGFP cells. Assessment of the 24 off-target sites for these four RGNs originally identified in U20S.EGFP cells revealed that many were again mutated in HEK293 and K562 cells with frequencies similar to those at their corresponding on-target site. As expected, DNA sequencing of a subset of these off-target sites from HEK293 cells provided additional molecular evidence that alterations are occurring at the expected genomic loci. It is not known for certain why in HEK293 cells four and in K562 cells eleven of the off-target sites identified in U20S.EGFP cells did not show detectable mutations. However, many of these off- target sites also showed relatively lower mutation frequencies in U20S.EGFP cells. Therefore, mutation rates of these sites in HEK293 and K562 cells may be falling below the reliable detection limit of our T7EI assay (-2-5%) because RGNs generally appear to have lower activities in HEK293 and K562 cells compared with
U20S.EGFP cells in our experiments. Taken together, the results in HEK293 and K562 cells provide evidence that the high-frequency off-target mutations we observe with RGNs will be a general phenomenon seen in multiple human cell types. Example le. Titration of gRNA- and Cas9-expressing plasmid amounts used for the EGFP disruption assay
Single guide RNAs (sgRNAs) were generated for three different sequences (EGFP SITES 1-3, shown above) located upstream of EGFP nucleotide 502, a position at which the introduction of frameshift mutations via non-homologous end- joining can robustly disrupt expression of EGFP (Maeder, M.L. et al, Mol Cell 31, 294-301 (2008); Reyon, D. et al, Nat Biotech 30, 460-465 (2012)).
For each of the three target sites, a range of gRNA-expressing plasmid amounts (12.5 to 250 ng) was initially trans fected together with 750 ng of a plasmid expressing a codon-optimized version of the Cas9 nuclease into our U20S.EGFP reporter cells bearing a single copy, constitutively expressed EGFP-PEST reporter gene. All three RGNs efficiently disrupted EGFP expression at the highest concentration of gRNA plasmid (250 ng) (Fig. 3E (top)). However, RGNs for target sites #1 and #3 exhibited equivalent levels of disruption when lower amounts of gRNA-expressing plasmid were transfected whereas RGN activity at target site #2 dropped immediately when the amount of gRNA-expressing plasmid transfected was decreased (Fig. 3E (top)).
The amount of Cas9-encoding plasmid (range from 50 ng to 750 ng) transfected into our U20S.EGFP reporter cells was titrated EGFP disruption assayed. As shown in Fig. 3F (top), target site #1 tolerated a three-fold decrease in the amount of Cas9-encoding plasmid transfected without substantial loss of EGFP disruption activity. However, the activities of RGNs targeting target sites #2 and #3 decreased immediately with a three-fold reduction in the amount of Cas9 plasmid transfected (Fig. 3F (top)). Based on these results, 25ng/250ng, 250ng/750ng, and 200ng/750ng of gRNA-/Cas9-expressing plasmids were used for EGFP target sites #1, #2, and #3, respectively, for the experiments described in Examples la- Id.
The reasons why some gRNA/Cas9 combinations work better than others in disrupting EGFP expression is not understood, nor is why some of these combinations are more or less sensitive to the amount of plasmids used for transfection. Although it is possible that the range of off-target sites present in the genome for these three sgRNAs might influence each of their activities, no differences were seen in the numbers of genomic sites that differ by one to six bps for each of these particular target sites (Table 1) that would account for the differential behavior of the three sgRNAs.
Table 1
Numbers of off-target sites in the human genome for six RGNs targeted to endo enous human genes and three RGNs tar eted to the EGFP re orter ene
Figure imgf000040_0001
Off-target sites for each of the six RGNs targeted to the VEGFA, RNF2, FANCF, and EMX1 genes and the three RGNs targeted to EGFP Target Sites #1, #2 and #3 were identified in human genome sequence build GRCh37. Mismatches were only allowed for the 20 nt region to which the gRNA anneals and not to the PAM sequence.
Example 2: Using pairs of guideRNAs with FokI-dCas9 fusion proteins
Monomeric CRISPR-Cas9 nucleases are widely used for targeted genome editing but can induce unwanted off-target mutations with high frequencies. This example describes new dimeric RNA-guided Fokl Nucleases (RFNs) that recognize an extended, double-length sequence and that strictly depend on two single guide RNAs (gRNAs) for cleavage activity. RFNs can robustly edit DNA sequences in endogenous human genes with high efficiencies. Additionally, a method for expressing gRNAs bearing any 5 ' end nucleotide is described, a critical advance that gives dimeric RFNs a useful targeting range. In direct comparisons, monomeric Cas9 nickases generally induce unwanted indels and unexpected focal point mutations with higher frequencies than RFNs directed by a matched single gRNA. RFNs combine the ease of CRISPR RNA-based targeting with the specificity enhancements of dimerization and provide an important new platform for research and therapeutic applications that require highly precise genome editing.
Materials and Methods
The following materials and methods were used in Example 2.
Single and multiplex gRNA expression plasmids
Plasmids encoding single or multiplex gRNAs were assembled in a single-step ligation of annealed target site oligosduplexes (Integrated DNA Technologies) and a constant region oligoduplex (for multiplex gRNAs) with BsmBI-digested Csy4- flanked gRNA backbone (pSQT1313; Addgene).
Multiplex gRNA encoding plasmids were constructed by ligating: 1) annealed oligos encoding the first target site, 2) phosphorylated annealed oligos encoding crRNA, tracrRNA, and Csy4-binding site, and 3) annealed oligos encoding the second target site, into a U6-Csy4site-gRNA plasmid backbone digested with BsmBI Type lis restriction enzyme. Csy4 RNA binding sites were attached to the 3' and 5' ends of a gRNA sequence and expressed with Cas9 in cells. The Csy4 RNA binding site sequence ' GUUC ACUGCCGUAUAGGC AGCUAAGAAA' (SEQ ID NO:20) was fused to the 5' and 3' end of the standard gRNA sequence.
GUUCACUGCCGUAUAGGCAGNNNNNNNNNNNNNNNNNNNNGUUUUAGAGCUA GAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUUGAAAAAGUGGCA CCGAGUCGGUGCGUUCACUGCCGUAUAGGCAGNNNNNNNNNNNNNNNNNNNN GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAGGCUAGUCCGUUAUCAACUU GAAAAAGUGGCACCGAGUCGGUGCGUUCACUGCCGUAUAGGCAG (SEQ ID NO: 21)
This sequence is a multiplex gRNA sequence flanked by Csy4 sites (underlined). Functionally, encoding these in multiplex on one transcript should have the same result as encoding them separately. Although all pairs of Csy4-flanked sgRNAs were expressed in a multiplex context in the experiments described herein, the sgRNAs can be encoded in multiplex sgRNAs separated by Csy4 sites encoded on one transcript as well as individual sgRNAs that have an additional Csy4 sequence. In this sequence, the first N20 sequence represents the sequence complementary to one strand of the target genomic sequence, and the second N20 sequence represents the sequence complementary to the other strand of the target genomic sequence.
A plasmid encoding the Csy4 recognition site containing gR A was co- transfected with plasmid encoding Cas9 and Csy4 proteins separated by a '2A' peptide linkage. The results showed that gR As with Csy4 sites fused to the 5' and 3 ' ends remained capable of directing Cas9-mediated cleavage in human cells using the U20S-EGFP disruption assay previously described. Thus, Csy4 R A binding sites can be attached to 3 ' end of a gRNA sequence and complexes of these Csy4 site- containing gRNAs with Cas9 remain functional in the cell.
In some experiments, a construct encoding Csy4-T2A-FokI-dCas9 was used.
The sequences of the FokI-dCas9 fusions are shown below, and include a GGGGS (SEQ ID NO:23) linker (underlined) between the Fokl and dCas9 and a nuclear localization sequence.
FokI-dCas9 amino acid sequence (FokI-G4S-dCas9-nls-3XFLAG) MQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNSTQDRILEMKVMEFFMKVYGYR GKHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMQRYVEENQTRN KHINPNEWWKVYPSSVTEFKFLFVSGHFKGNYKAQLTRLNHITNCNGAVLSVEELLI GGEMIKAGTLTLEEVRRKFNNGEINFGGGGSDKKYS IGLAIGTNSVGWAVITDEYKV PSKKFKVLGN DRHS IKKNLIGALLFDSGE AEATRLKRTARRRYTRRKNRICYLQE IFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFG IVDEVAYHEKYP IYHLRKK LVDS DKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEE NPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSN FDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEI TKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQ EEFYKFIKPILEKMDGTEELLVKLNREDLLRKQR FDNGS I PHQIHLGELHAILRRQ EDFYPFLKDNREKIEKIL FRI PYYVGPLARGNSRFAWMTRKSEE I PWNFEEVVD KGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFL SGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEI SGVEDRFNASLGTYHDL LKI IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRR RYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKA QVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQ TTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVD QELDINRLSDYDVDAIVPQSFLKDDS IDNKVLTRSDKNRGKSDNVPSEEVVKKMKNY WRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRM NTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTA LIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEI LA NGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESI LPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGIT IMERSSFEKNPIDFLEAKGYKEVKKDLI IKLPKYSLFELENGRKRMLASAGELQKGN ELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEI IEQI SEFSKRVI LADANLDKVLSAYNKHRDKPIREQAE I IHLFTLTNLGAPAAFKYFD IDRKRY S TKEVLDATLIHQSI GLYETRIDLSQLGGDGSPKKKRKVSSDYKDHDGDYKDHDIDY KDDDDK (SEQ ID NO:24)
FokI-dCas9 nucleotide sequence (FokI-G4S-dCas9-nls-3XFLAG)
ATGCAACTAGTCAAAAGTGAACTGGAGGAGAAGAAATCTGAACTTCGTCATAAATTG AAATATGTGCCTCATGAATATATTGAATTAATTGAAATTGCCAGAAATTCCACTCAG GATAGAATTCTTGAAATGAAGGTAATGGAATTTTTTATGAAAGTTTATGGATATAGA GGTAAACATTTGGGTGGATCAAGGAAACCGGACGGAGCAATTTATACTGTCGGATCT CCTATTGATTACGGTGTGATCGTGGATACTAAAGCTTATAGCGGAGGTTATAATCTG CCAATTGGCCAAGCAGATGAAATGCAACGATATGTCGAAGAAAATCAAACACGAAAC AAACATATCAACCCTAATGAATGGTGGAAAGTCTATCCATCTTCTGTAACGGAATTT AAGTTTTTATTTGTGAGTGGTCACTTTAAAGGAAACTACAAAGCTCAGCTTACACGA TTAAATCATATCACTAATTGTAATGGAGCTGTTCTTAGTGTAGAAGAGCTTTTAATT GGTGGAGAAATGATTAAAGCCGGCACATTAACCTTAGAGGAAGTCAGACGGAAATTT AATAACGGCGAGATAAACTTTGGTGGCGGTGGATCCGATAAAAAGTATTCTATTGGT TTAGCCATCGGCACTAATTCCGTTGGATGGGCTGTCATAACCGATGAATACAAAGTA CCTTCAAAGAAATTTAAGGTGTTGGGGAACACAGACCGTCATTCGATTAAAAAGAAT CTTATCGGTGCCCTCCTATTCGATAGTGGCGAAACGGCAGAGGCGACTCGCCTGAAA CGAACCGCTCGGAGAAGGTATACACGTCGCAAGAACCGAATATGTTACTTACAAGAA ATTTTTAGCAATGAGATGGCCAAAGTTGACGATTCTTTCTTTCACCGTTTGGAAGAG TCCTTCCTTGTCGAAGAGGACAAGAAACATGAACGGCACCCCATCTTTGGAAACATA GTAGATGAGGTGGCATATCATGAAAAGTACCCAACGATTTATCACCTCAGAAAAAAG CTAGTTGACTCAACTGATAAAGCGGACCTGAGGTTAATCTACTTGGCTCTTGCCCAT ATGATAAAGTTCCGTGGGCACTTTCTCATTGAGGGTGATCTAAATCCGGACAACTCG GATGTCGACAAACTGTTCATCCAGTTAGTACAAACCTATAATCAGTTGTTTGAAGAG AACCCTATAAATGCAAGTGGCGTGGATGCGAAGGCTATTCTTAGCGCCCGCCTCTCT AAATCCCGACGGCTAGAAAACCTGATCGCACAATTACCCGGAGAGAAGAAAAATGGG TTGTTCGGTAACCTTATAGCGCTCTCACTAGGCCTGACACCAAATTTTAAGTCGAAC TTCGACTTAGCTGAAGATGCCAAATTGCAGCTTAGTAAGGACACGTACGATGACGAT CTCGACAATCTACTGGCACAAATTGGAGATCAGTATGCGGACTTATTTTTGGCTGCC AAAAACCTTAGCGATGCAATCCTCCTATCTGACATACTGAGAGTTAATACTGAGATT ACCAAGGCGCCGTTATCCGCTTCAATGATCAAAAGGTACGATGAACATCACCAAGAC TTGACACTTCTCAAGGCCCTAGTCCGTCAGCAACTGCCTGAGAAATATAAGGAAATA TTCTTTGATCAGTCGAAAAACGGGTACGCAGGTTATATTGACGGCGGAGCGAGTCAA GAGGAATTCTACAAGTTTATCAAACCCATATTAGAGAAGATGGATGGGACGGAAGAG TTGCTTGTAAAACTCAATCGCGAAGATCTACTGCGAAAGCAGCGGACTTTCGACAAC GGTAGCATTCCACATCAAATCCACTTAGGCGAATTGCATGCTATACTTAGAAGGCAG GAGGATTTTTATCCGTTCCTCAAAGACAATCGTGAAAAGATTGAGAAAATCCTAACC TTTCGCATACCTTACTATGTGGGACCCCTGGCCCGAGGGAACTCTCGGTTCGCATGG ATGACAAGAAAGTCCGAAGAAACGAT ACTCCATGGAATTTTGAGGAAGTTGTCGAT AAAGGTGCGTCAGCTCAATCGTTCATCGAGAGGATGACCAACTTTGACAAGAATTTA CCGAACGAAAAAGTATTGCCTAAGCACAGTTTACTTTACGAGTATTTCACAGTGTAC AATGAACTCACGAAAGTTAAGTATGTCACTGAGGGCATGCGTAAACCCGCCTTTCTA AGCGGAGAACAGAAGAAAGCAATAGTAGATCTGTTATTCAAGACCAACCGCAAAGTG ACAGTTAAGCAATTGAAAGAGGACTACTTTAAGAAAATTGAATGCTTCGATTCTGTC GAGATCTCCGGGGTAGAAGATCGATTTAATGCGTCACTTGGTACGTATCATGACCTC CTAAAGATAATTAAAGATAAGGACTTCCTGGATAACGAAGAGAATGAAGATATCTTA GAAGATATAGTGTTGACTCTTACCCTCTTTGAAGATCGGGAAATGATTGAGGAAAGA CTAAAAACATACGCTCACCTGTTCGACGATAAGGTTATGAAACAGTTAAAGAGGCGT CGCTATACGGGCTGGGGACGATTGTCGCGGAAACTTATCAACGGGATAAGAGACAAG CAAAGTGGTAAAACTATTCTCGATTTTCTAAAGAGCGACGGCTTCGCCAATAGGAAC TTTATGCAGCTGATCCATGATGACTCTTTAACCTTCAAAGAGGATATACAAAAGGCA CAGGTTTCCGGACAAGGGGACTCATTGCACGAACATATTGCGAATCTTGCTGGTTCG CCAGCCATCAAAAAGGGCATACTCCAGACAGTCAAAGTAGTGGATGAGCTAGTTAAG GTCATGGGACGTCACAAACCGGAAAACATTGTAATCGAGATGGCACGCGAAAATCAA ACGACTCAGAAGGGGCAAAAAAACAGTCGAGAGCGGATGAAGAGAATAGAAGAGGGT ATTAAAGAACTGGGCAGCCAGATCTTAAAGGAGCATCCTGTGGAAAATACCCAATTG CAGAACGAGAAACTTTACCTCTATTACCTACAAAATGGAAGGGACATGTATGTTGAT CAGGAACTGGACATAAACCGTTTATCTGATTACGACGTCGATGCCATTGTACCCCAA TCCTTTTTGAAGGACGATTCAATCGACAATAAAGTGCTTACACGCTCGGATAAGAAC CGAGGGAAAAGTGACAATGTTCCAAGCGAGGAAGTCGTAAAGAAAATGAAGAACTAT TGGCGGCAGCTCCTAAATGCGAAACTGATAACGCAAAGAAAGTTCGATAACTTAACT AAAGCTGAGAGGGGTGGCTTGTCTGAACTTGACAAGGCCGGATTTATTAAACGTCAG CTCGTGGAAACCCGCCAAATCACAAAGCATGTTGCACAGATACTAGATTCCCGAATG AATACGAAATACGACGAGAACGATAAGCTGATTCGGGAAGTCAAAGTAATCACTTTA AAGTCAAAATTGGTGTCGGACTTCAGAAAGGATTTTCAATTCTATAAAGTTAGGGAG ATAAATAACTACCACCATGCGCACGACGCTTATCTTAATGCCGTCGTAGGGACCGCA CTCATTAAGAAATACCCGAAGCTAGAAAGTGAGTTTGTGTATGGTGATTACAAAGTT TATGACGTCCGTAAGATGATCGCGAAAAGCGAACAGGAGATAGGCAAGGCTACAGCC AAATACTTCTTTTATTCTAACATTATGAATTTCTTTAAGACGGAAATCACTCTGGCA AACGGAGAGATACGCAAACGACCTTTAATTGAAACCAATGGGGAGACAGGTGAAATC GTATGGGATAAGGGCCGGGACTTCGCGACGGTGAGAAAAGTTTTGTCCATGCCCCAA GTCAACATAGTAAAGAAAACTGAGGTGCAGACCGGAGGGTTTTCAAAGGAATCGATT CTTCCAAAAAGGAATAGTGATAAGCTCATCGCTCGTAAAAAGGACTGGGACCCGAAA AAGTACGGTGGCTTCGATAGCCCTACAGTTGCCTATTCTGTCCTAGTAGTGGCAAAA GTTGAGAAGGGAAAATCCAAGAAACTGAAGTCAGTCAAAGAATTATTGGGGATAACG ATTATGGAGCGCTCGTCTTTTGAAAAGAACCCCATCGACTTCCTTGAGGCGAAAGGT TACAAGGAAGTAAAAAAGGATCTCATAATTAAACTACCAAAGTATAGTCTGTTTGAG TTAGAAAATGGCCGAAAACGGATGTTGGCTAGCGCCGGAGAGCTTCAAAAGGGGAAC GAACTCGCACTACCGTCTAAATACGTGAATTTCCTGTATTTAGCGTCCCATTACGAG AAGTTGAAAGGTTCACCTGAAGATAACGAACAGAAGCAACTTTTTGTTGAGCAGCAC AAACATTATCTCGACGAAATCATAGAGCAAATTTCGGAATTCAGTAAGAGAGTCATC CTAGCTGATGCCAATCTGGACAAAGTATTAAGCGCATACAACAAGCACAGGGATAAA CCCATACGTGAGCAGGCGGAAAATATTATCCATTTGTTTACTCTTACCAACCTCGGC GCTCCAGCCGCATTCAAGTATTTTGACACAACGATAGATCGCAAACGATACACTTCT ACCAAGGAGGTGCTAGACGCGACACTGATTCACCAATCCATCACGGGATTATATGAA ACTCGGATAGATTTGTCACAGCTTGGGGGTGACGGATCCCCCAAGAAGAAGAGGAAA GTCTCGAGCGACTACAAAGACCATGACGGTGATTATAAAGATCATGACATCGATTAC AAGGATGACGATGACAAGTGA (SEQ ID NO: 25)
Alternatively, a human codon optimized version of the construct was used, which contained both N- and C-terminal nuclear localization signals, as shown below.
Nls-FokI-dCas9-nls amino acid sequence
MPKKKRKVSSQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNSTQDRILEMKVME FFMKVYGYRGKHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMQR YVEENQTRNKHINPNEWWKVYPSSVTEFKFLFVSGHFKGNYKAQLTRLNHITNCNGA VLSVEELLIGGEMIKAGTLTLEEVRRKFNNGEINFGGGGSDKKYS IGLAIGTNSVGW AVI DEYKVPSKKFKVLGN DRHS IKKNLIGALLFDSGE AEATRLKRTARRRYTRR KNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKY P IYHLRKKLVDS DKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLV QTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSL GLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLS DILRVNTEI KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYA GYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQR FDNGS I PHQIHLG ELHAILRRQEDFYPFLKDNREKIEKIL FRI PYYVGPLARGNSRFAWMTRKSEE I PWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVT EGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEI SGVEDRFN ASLGTYHDLLKI IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDD KVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSL TFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYL QNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDDS IDNKVLTRSDKNRGKSDNVPSE EVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA YLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMN FFKTEI LANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQ TGGFSKES ILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLK SVKELLGI IMERSSFEKNPIDFLEAKGYKEVKKDLI IKLPKYSLFELENGRKRMLA SAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQ I SEFSKRVILADANLDKVLSAYNKHRDKPIREQAENI IHLFTLTNLGAPAAFKYFDT IDRKRYTSTKEVLDATLIHQSI GLYETRIDLSQLGGDGSPKKKRKVSSDYKDHDG DYKDHDIDYKDDDDK (SEQ ID NO: 26)
Nls-Fokl— dCas9-nls nucleotide sequence
ATGCCTAAGAAGAAGCGGAAGGTGAGCAGCCAACTTGTGAAGTCTGAACTCGAGGAG AAAAAATCAGAGTTGAGACACAAGTTGAAGTACGTGCCACACGAATACATCGAGCTT ATCGAGATCGCCAGAAACAGTACCCAGGATAGGATCCTTGAGATGAAAGTCATGGAG TTCTTTATGAAGGTCTACGGTTATAGAGGAAAGCACCTTGGCGGTAGCAGAAAGCCC GATGGCGCCATCTATACTGTCGGATCTCCTATCGATTATGGGGTGATCGTGGATACC AAAGCTTACTCAGGCGGGTACAACTTGCCCATAGGACAAGCCGACGAGATGCAGCGG TATGTCGAAGAGAACCAGACGCGCAACAAGCACATCAACCCCAATGAATGGTGGAAA GTGTACCCAAGTAGTGTGACTGAGTTCAAGTTCCTGTTTGTCTCCGGCCACTTTAAG GGCAATTATAAAGCTCAGCTCACTAGACTCAATCACATCACAAACTGCAACGGAGCT GTGTTGTCAGTGGAGGAGCTCCTGATTGGAGGCGAGATGATCAAAGCCGGCACCCTT ACACTGGAGGAGGTGCGGCGGAAGTTCAACAATGGAGAGATCAACTTCGGTGGCGGT GGATCCGATAAAAAGTATTCTATTGGTTTAGCCATCGGCACTAATTCCGTTGGATGG GCTGTCATAACCGATGAATACAAAGTACCTTCAAAGAAATTTAAGGTGTTGGGGAAC ACAGACCGTCATTCGATTAAAAAGAATCTTATCGGTGCCCTCCTATTCGATAGTGGC GAAACGGCAGAGGCGACTCGCCTGAAACGAACCGCTCGGAGAAGGTATACACGTCGC AAGAACCGAATATGTTACTTACAAGAAATTTTTAGCAATGAGATGGCCAAAGTTGAC GATTCTTTCTTTCACCGTTTGGAAGAGTCCTTCCTTGTCGAAGAGGACAAGAAACAT GAACGGCACCCCATCTTTGGAAACATAGTAGATGAGGTGGCATATCATGAAAAGTAC CCAACGATTTATCACCTCAGAAAAAAGCTAGTTGACTCAACTGATAAAGCGGACCTG AGGTTAATCTACTTGGCTCTTGCCCATATGATAAAGTTCCGTGGGCACTTTCTCATT GAGGGTGATCTAAATCCGGACAACTCGGATGTCGACAAACTGTTCATCCAGTTAGTA CAAACCTATAATCAGTTGTTTGAAGAGAACCCTATAAATGCAAGTGGCGTGGATGCG AAGGCTATTCTTAGCGCCCGCCTCTCTAAATCCCGACGGCTAGAAAACCTGATCGCA CAATTACCCGGAGAGAAGAAAAATGGGTTGTTCGGTAACCTTATAGCGCTCTCACTA GGCCTGACACCAAATTTTAAGTCGAACTTCGACTTAGCTGAAGATGCCAAATTGCAG CTTAGTAAGGACACGTACGATGACGATCTCGACAATCTACTGGCACAAATTGGAGAT CAGTATGCGGACTTATTTTTGGCTGCCAAAAACCTTAGCGATGCAATCCTCCTATCT GACATACTGAGAGTTAATACTGAGATTACCAAGGCGCCGTTATCCGCTTCAATGATC AAAAGGTACGATGAACATCACCAAGACTTGACACTTCTCAAGGCCCTAGTCCGTCAG CAACTGCCTGAGAAATATAAGGAAATATTCTTTGATCAGTCGAAAAACGGGTACGCA GGTTATATTGACGGCGGAGCGAGTCAAGAGGAATTCTACAAGTTTATCAAACCCATA TTAGAGAAGATGGATGGGACGGAAGAGTTGCTTGTAAAACTCAATCGCGAAGATCTA CTGCGAAAGCAGCGGACTTTCGACAACGGTAGCATTCCACATCAAATCCACTTAGGC GAATTGCATGCTATACTTAGAAGGCAGGAGGATTTTTATCCGTTCCTCAAAGACAAT CGTGAAAAGATTGAGAAAATCCTAACCTTTCGCATACCTTACTATGTGGGACCCCTG GCCCGAGGGAACTCTCGGTTCGCATGGATGACAAGAAAGTCCGAAGAAACGATTACT CCATGGAATTTTGAGGAAGTTGTCGATAAAGGTGCGTCAGCTCAATCGTTCATCGAG AGGATGACCAACTTTGACAAGAATTTACCGAACGAAAAAGTATTGCCTAAGCACAGT TTACTTTACGAGTATTTCACAGTGTACAATGAACTCACGAAAGTTAAGTATGTCACT GAGGGCATGCGTAAACCCGCCTTTCTAAGCGGAGAACAGAAGAAAGCAATAGTAGAT CTGTTATTCAAGACCAACCGCAAAGTGACAGTTAAGCAATTGAAAGAGGACTACTTT AAGAAAATTGAATGCTTCGATTCTGTCGAGATCTCCGGGGTAGAAGATCGATTTAAT GCGTCACTTGGTACGTATCATGACCTCCTAAAGATAATTAAAGATAAGGACTTCCTG GATAACGAAGAGAATGAAGATATCTTAGAAGATATAGTGTTGACTCTTACCCTCTTT GAAGATCGGGAAATGATTGAGGAAAGACTAAAAACATACGCTCACCTGTTCGACGAT AAGGTTATGAAACAGTTAAAGAGGCGTCGCTATACGGGCTGGGGACGATTGTCGCGG AAACTTATCAACGGGATAAGAGACAAGCAAAGTGGTAAAACTATTCTCGATTTTCTA AAGAGCGACGGCTTCGCCAATAGGAACTTTATGCAGCTGATCCATGATGACTCTTTA ACCTTCAAAGAGGATATACAAAAGGCACAGGTTTCCGGACAAGGGGACTCATTGCAC GAACATATTGCGAATCTTGCTGGTTCGCCAGCCATCAAAAAGGGCATACTCCAGACA GTCAAAGTAGTGGATGAGCTAGTTAAGGTCATGGGACGTCACAAACCGGAAAACATT GTAATCGAGATGGCACGCGAAAATCAAACGACTCAGAAGGGGCAAAAAAACAGTCGA GAGCGGATGAAGAGAATAGAAGAGGGTATTAAAGAACTGGGCAGCCAGATCTTAAAG GAGCATCCTGTGGAAAATACCCAATTGCAGAACGAGAAACTTTACCTCTATTACCTA CAAAATGGAAGGGACATGTATGTTGATCAGGAACTGGACATAAACCGTTTATCTGAT TACGACGTCGATGCCATTGTACCCCAATCCTTTTTGAAGGACGATTCAATCGACAAT AAAGTGCTTACACGCTCGGATAAGAACCGAGGGAAAAGTGACAATGTTCCAAGCGAG GAAGTCGTAAAGAAAATGAAGAACTATTGGCGGCAGCTCCTAAATGCGAAACTGATA AC G C AAAG AAAG T T C G AT AAC T T AAC T AAAG CTGAGAGGGGTGGCTTGTCT G AAC T T GACAAGGCCGGATTTATTAAACGTCAGCTCGTGGAAACCCGCCAAATCACAAAGCAT GTTGCACAGATACTAGATTCCCGAATGAATACGAAATACGACGAGAACGATAAGCTG ATTCGGGAAGTCAAAGTAATCACTTTAAAGTCAAAATTGGTGTCGGACTTCAGAAAG GATTTTCAATTCTATAAAGTTAGGGAGATAAATAACTACCACCATGCGCACGACGCT TATCTTAATGCCGTCGTAGGGACCGCACTCATTAAGAAATACCCGAAGCTAGAAAGT GAGTTTGTGTATGGTGATTACAAAGTTTATGACGTCCGTAAGATGATCGCGAAAAGC GAACAGGAGATAGGCAAGGCTACAGCCAAATACTTCTTTTATTCTAACATTATGAAT TTCTTTAAGACGGAAATCACTCTGGCAAACGGAGAGATACGCAAACGACCTTTAATT GAAACCAATGGGGAGACAGGTGAAATCGTATGGGATAAGGGCCGGGACTTCGCGACG GTGAGAAAAGTTTTGTCCATGCCCCAAGTCAACATAGTAAAGAAAACTGAGGTGCAG ACCGGAGGGTTTTCAAAGGAATCGATTCTTCCAAAAAGGAATAGTGATAAGCTCATC GCTCGTAAAAAGGACTGGGACCCGAAAAAGTACGGTGGCTTCGATAGCCCTACAGTT GCCTATTCTGTCCTAGTAGTG G C AAAAG T T G AG AAG G G AAAAT C C AAG AAAC T G AAG TCAGTCAAAGAATTATTGGGGATAACGATTATGGAGCGCTCGTCTTTTGAAAAGAAC CCCATCGACTTCCTTGAGGCGAAAGGTTACAAGGAAGTAAAAAAGGATCTCATAATT AAACTACCAAAGTATAGTCTGTTTGAGTTAGAAAATGGCCGAAAACGGATGTTGGCT AGCGCCGGAGAGCTTCAAAAGGGGAACGAACTCGCACTACCGTCTAAATACGTGAAT TTCCTGTATTTAGCGTCCCATTACGAGAAGTTGAAAGGTTCACCTGAAGATAACGAA CAGAAGCAACTTTTTGTTGAGCAGCACAAACATTATCTCGACGAAATCATAGAGCAA ATTTCGGAATTCAGTAAGAGAGTCATCCTAGCTGATGCCAATCTGGACAAAGTATTA AGCGCATACAACAAGCACAGGGATAAACCCATACGTGAGCAGGCGGAAAATATTATC CATTTGTTTACTCTTACCAACCTCGGCGCTCCAGCCGCATTCAAGTATTTTGACACA ACGATAGATCGCAAACGATACACTTCTACCAAGGAGGTGCTAGACGCGACACTGATT CACCAATCCATCACGGGATTATATGAAACTCGGATAGATTTGTCACAGCTTGGGGGT GACGGATCCCCCAAGAAGAAGAGGAAAGTCTCGAGCGACTACAAAGACCATGACGGT GA A AAAGA CA GACA CGA ACAAGGA GACGA GACAAG GA (SEQ ID NO: 27) Tissue culture and transfections
All cell culture experiments were carried out in HEK 293 cells, U20S cells, or in U20S cells harboring a stably integrated, single-copy, destabilized EGFP gene (U20S.EGFP cells). Cell lines were cultured in Advanced DMEM (Life
Technologies) supplemented with 10% FBS, 2 mM GlutaMax (Life Technologies) and penicillin/streptomycin at 37C with 5% C02. Additionally, U20S.EGFP cells were cultured in the presence of 400 μg/ml of G418.
U20S cells and U20S.EGFP cells were transfected using the DN-100 program of a Lonza 4D-Nucleofector according to the manufacturer's instructions. In initial FokI-dCas9 activity screens and focused spacer length analysis experiments, 750 ng of pCAG-Csy4-FokI-dCas9-nls nuclease plasmid and 250 ng of gRNA encoding plasmids were transfected together with 50 ng tdTomato expression plasmid (Clontech) as a transfection control. In all other experiments in U20S and
U20S.EGFP cells, 975 ng of human codon optimized pCAG-Csy4-T2A-nls-hFokI- dCas9-nls (SQT1601) or pCAG-Cas9-D10A nickase (NW3) were transfected along with 325 ng of gRNA vector and 10 ng of Td tomato expression plasmid and analyzed 3 days after transfection. HEK293 cells were transfected with 750 ng of nuclease plasmid, 250 ng of gRNA expression plasmid and 10 ng of Td tomato, using
Lipofectamine (Life Technologies) according to the manufacturer's instructions and analyzed for NHEJ-mediated mutagenesis 3 days after transfection.
Single transfections were performed for the initial spacer activity screen, and duplicate transfections for the focused spacer length analysis. All other transfections were performed in triplicate. EGFP disruption assay
The EGFP disruption assay was performed as previously described (see Example 1 and Reyon et al, Nat Biotech 30, 460-465 (2012)) using U20S.EGFP reporter cells. Cells were assayed for EGFP and tdTomato expression using an BD Biosciences LSR II or Fortessa FACS analyzer.
Quantification of nuclease- or nickase-induced mutation rates by T7EI assay
T7E1 assays were performed as previously described (Reyon et al, Nat Biotech 30, 460-465 (2012)). Briefly, genomic DNA was isolated 72 hours post transfection using the Agencourt DNAdvance Genomic DNA Isolation kit (Beckman Coulter Genomics) according to the manufacturer's instructions with a Sciclone G3 liquid-handling workstation (Caliper). PCR reactions to amplify genomic loci were performed using Phusion Hot- start Flex DNA polymerase (New England Biolabs). Samples were amplified using a two-step protocol (98 °C, 30 sec; (98 °C, 7 sec; 72 °C, 30 sec) x 35; 72 °C, 5 min) or a touchdown PCR protocol ((98 °C, 10 s; 72-62 °C, -1 °C/cycle, 15 s; 72 °C, 30 s) 10 cycles, (98 °C, 10 s; 62 °C, 15 s; 72 °C, 30 s) 25 cycles). 200 ng of purified PCR amplicons were denatured, hybridized, and treated with T7 Endonuclease I (New England Biolabs). Mutation frequency was quantified using a Qiaxcel capillary electrophoresis instrument (Qiagen) as previously described (Reyon et al, Nat Biotech 30, 460-465 (2012)).
Sanger sequencing of mutagenized genomic DNA
The same purified PCR products used for T7EI assay were Topo-cloned (Life Technologies) and plasmid DNA of individual clones was isolated and sequenced using an M13 reverse primer (5'-GTAAAACGACGGCCAG-3'; SEQ ID NO: 19).
Illumina Library Preparation and Analysis
Short 200-350 bp PCR products were amplified using Phusion Hot- start FLEX DNA polymerase. PCR products were purified using Ampure XP beads (Beckman Coulter Genomics) according to manufacturer's instructions. Dual-indexed TruSeq Illumina deep sequencing libraries were prepared using a high-throughput library preparation system (Kapa Biosystems) on a Sciclone G3 liquid-handling workstation. Final adapter-ligated libraries were quantified using a Qiaxcel capillary electrophoresis instrument (Qiagen). 150 bp paired end sequencing was performed on an Illumina MiSeq Sequencer by the Dana-Farber Cancer Institute Molecular Biology Core.
MiSeq paired-end reads were mapped to human genome reference GChr37 using bwa. Reads with an average quality score >30 were analyzed for insertion or deletion mutations that overlapped the intended target or candidate off-target nuclease binding site. Mutation analyses were conducted using the Genome Analysis Toolkit (GATK) and Python.
Off-target search algorithm:
A target-site matching algorithm was implemented that looks for matches with less than a specified number of mismatches in a sliding window across the human genome.
Example 2a. Rationale for designing dimeric RNA-guided nucleases
It was hypothesized that a single platform combining the specificity advantages of dimerization with the ease of Cas9 targeting could be developed. To do this, the well-characterized, dimerization-dependent Fokl nuclease domain was fused to a RNA-guided catalytically inactive Cas9 (dCas9) protein. It was hoped that, like Fokl-containing ZFNs and TALENs, dimers of these fusions might mediate sequence- specific DNA cleavage when bound to target sites composed of two "half-sites" with a certain length "spacer" sequence between them (Fig. 4A). Such fusions were hypothesized to have enhanced specificity because they should require two gRNAs for activity (Fig. 4A) and because a single gRNA would presumably be too inefficient or unable to recruit the two Fokl-containing fusion proteins required for DNA cleavage. It was hypothesized that such a dimeric system would show improved specificity relative to standard monomeric Cas9 nucleases and also would potentially possess important specificity advantages over the paired nickase system in which single nickases can still exert unwanted mutagenic effects.
Example 2b. Multiplex expression of gRNAs without 5 '-end nucleotide limitations
The targeting range for a dimeric RNA-guided nuclease would be low using existing gRNA expression methods. Two sequence requirements typically restrict the targeting range of a dCas9 monomer: the requirement for a PAM sequence of 5'- NGG that is specified by the dCas9 and a requirement for a G nucleotide at the 5' end of the gRNA imposed by the use of a U6 promoter in most expression vectors. If, however, the requirement for the 5 ' G in the gRNA could be relieved, then the targeting range would improve by 16-fold.
To develop a multiplex system that would allow for the expression of gRNAs with any 5' nucleotide, a plasmid was constructed from which two gRNAs, each flanked by cleavage sites for the Csy4 ribonuclease (Haurwitz et al, Science 329, 1355-1358 (2010)), can be expressed within a single RNA transcribed from a U6 promoter (Fig. 4B). Csy4 would be expected to process this transcript thereby releasing the two gRNAs. Based on the known mechanism of Csy4-mediated cleavage ((Haurwitz et al, Science 329, 1355-1358 (2010); Sternberg et al, RNA 18, 661-672 (2012)), each processed gRNA should retain a Csy4 recognition site on its 3' end with a Csy4 protein bound to that site (Fig. 4B). In this configuration, it should be possible to express gRNAs with any 5' nucleotide. This system was tested by using it to express two gRNAs targeted to sites within the EGFP reporter gene. Co- expression of this transcript together with Csy4 and Cas9 nucleases in human cells led to the introduction of indel mutations at both EGFP target sites as well as of deletion of the sequence between these sites (Fig. 4C). These experiments suggest that both gRNAs are being processed from the single parental RNA transcript and both are capable of directing Cas9 nuclease activities in human cells.
Example 2c. Construction and optimization of dimeric RNA-guided nucleases Two different hybrid proteins harboring the Fokl nuclease domain and the dCas9 protein were constructed: one in which the Fokl nuclease domain is fused to the carboxy-terminus of dCas9 (dCas9-FokI) and the other in which it is fused to the amino-terminus (FokI-dCas9) (Fig. 5A). The dCas9-FokI protein is analogous in architecture to ZFNs and TALENs (Fig. 5A). To ascertain whether either or both of these fusions could mediate site-specific cleavage of DNA, a well-established human cell-based assay that can rapidly and easily quantify the introduction of NHEJ- mediated indels into an EGFP reporter gene was used (the EGFP disruption assay described above in Example 1). Because the geometry of the half- sites required for efficient cleavage was not known, 60 pairs of gRNAs targeted to various sites in EGFP were designed. The two half-sites targeted by each of these gRNA pairs were oriented such that both of their PAM sequences are either directly adjacent to the spacer sequence (the "PAM in" orientation) or positioned at the outer boundaries of the full-length target site (the "PAM out" orientation) (Fig. 5B). In addition, the spacer sequence was also varied in length from 0 to 31 bps (Fig. 5B and Table 2).
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Surprisingly, the dCas9-FokI protein did not show detectable EGFP disruption activity when co-expressed with any of the 60 gRNA pairs in human U20S.EGFP cells (Fig. 5E). However, screening of the FokI-dCas9 protein with the same 60 gRNA pairs did reveal EGFP disruption activity on target sites composed of half-sites in the PAM out orientation and with spacer lengths of 13 to 17 bps and of 26 bps (approximately one turn of the DNA helix more than the 13-17 bp spacer lengths) (Fig. 5B). Testing of FokI-dCas9 on an additional 25 target DNA sites with spacer lengths ranging from 10 to 20 bps and with half-sites in the PAM out orientation demonstrated efficient cleavage on targets with spacer lengths of 13 to 18 bps (Figs. 5C-D). In these experiments, one site each was tested for spacer lengths of 17 or 18 bps and not all sites with a 13 bp spacer length showed activity. Analysis of a subset of successfully targeted sites by T7EI analysis and Sanger sequencing further confirmed the presence of indels at the intended location. Thus FokI-dCas9 can be directed by two appropriately positioned gRNAs to efficiently cleave a full-length target site of interest. For simplicity, the complex of two FokI-dCas9 fusions and two gRNAs are referred to herein as RNA-guided Fokl Nucleases (RFNs).
To extend the initial findings with the EGFP reporter gene and to ascertain whether RFNs could be used to perform routine genome editing of endogenous human genes, gRNA pairs were designed for 12 different target sites in nine different human genes (Table 2). Eleven of the 12 RFNs tested introduced indels with high efficiencies (range of 3 to 40%) at their intended target sites in human U20S.EGFP cells as judged by T7EI assay (Table 2). Similar results were obtained with these same 12 RFN pairs in HEK293 cells (Table 2). Sanger sequencing of successfully targeted alleles from U20S.EGFP cells revealed the introduction of a range of indels (primarily deletions) at the expected cleavage site (Fig. 5F). The high success rate and high efficiencies of modifications observed in two different human cell lines demonstrate the robustness of RFNs for modifying endogenous human genes.
Example 2d. RFNs possess extended specificities for their cleavage sites To test whether RFNs possess enhanced recognition specificities associated with dimerization, whether these nucleases strictly depend upon the presence of both gRNAs in a pair was examined. In an ideal dimeric system, single gRNAs should not be able to efficiently direct FokI-dCas9-induced indels. To perform an initial test, two pairs of gRNAs directed to two target sites in EGFP were used that had been shown to efficiently direct FokI-dCas9-induced indels to their target sites (EGFP sites 47 and 81) in human U20S.EGFP cells (Fig. 5C). Replacement of one or the other gRNA in each of these two pairs with a gRNA targeted to an unrelated site in VEGFA resulted in reduction of EGFP disruption activity (Fig. 6A) and reduction of targeted mutations to undetectable levels as judged by T7EI assays (Fig. 6B). Similarly, the effects of using only one of each of the two gRNAs were tested using pairs that efficiently introduce FokI-dCas9-mediated indels in the human APC, MLH1, and VEGFA genes (Table 2) and again observed loss of detectable RFN-induced indels by T7EI assay (Fig. 6C). These results demonstrate that efficient induction of genome editing by an RFN requires two gRNAs with appropriate complementarity to the full- length target site.
Given that the activities of our RFNs depend on the expression of two gRNAs, it was hoped that their mutagenic effects on known off-target sites of one of the single gRNAs in the pair should be negligible. Performing these direct comparisons requires knowing the off-target sites for a monomeric Cas9 nuclease guided by a single gRNA that itself can also serve one of the two gRNAs needed to target a dimeric RFN.
Although very few monomeric Cas9 nuclease off-target sites have been defined in the literature, five off-target sites had been previously identified for one of the gRNAs we used to target the dimeric RFN site in the human VEGFA gene (Example 1). Deep sequencing was used to ascertain whether these five off-target sites showed evidence of mutations in cells in which the FEG ^-targeted RFNs had been expressed (these are the same cells we used for the T7EI assay shown in Fig. 6C). The frequency of indel mutations at all five off-target sites was indistinguishable from background (Fig. 6D and Table 3). These results demonstrate that the use of RFNs can essentially eliminate the off-target effects originally induced by Cas9 nuclease and a single gRNA and are consistent with our observation that a single gRNA expressed with FokI-dCas9 does not efficiently induce indels. Although, at present, it is not possible to perform these direct comparisons on additional sites - such experiments will have to await the identification of off-target sites for more single gRNA sites that can also target a half-site for a dimeric RFN, it was concluded that dimeric RFNs have enhanced specificities relative to standard monomeric Cas9 nucleases.
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Example 2e. Monomeric Cas9 nickases induce higher rates of mutagenesis than single gRNA/FokI-dCas9 complexes
As noted above, an important weakness of the paired Cas9 nickase approach is that single monomeric nickases can introduce indel mutations with high frequencies at certain target sites (See Example 1 and Ran et al, Cell 154, 1380-1389 (2013); Mali et al, Nat Biotechnol 31 , 833-838 (2013); Cho et al, Genome Res (2013); and Mali et al, Science 339, 823-826 (2013)). This lack of dimerization-dependence in the paired Cas9 nickase system is a potential source of off-target effects because the two monomeric nickases can each create unwanted indel mutations elsewhere in the genome. It was hypothesized that because RFNs introduce alterations using a dimerization-dependent Fokl nuclease, these fusions should generally show less undesirable indel activity in the presence of only one gRNA compared to what is observed with monomeric Cas9 nickases.
To test this hypothesis, the activities of FokI-dCas9 and Cas9 nickase were compared in the presence of a single gRNA at six dimeric human gene target sites (a total of 12 half-sites; Table 4). These particular sites were chosen because monomeric Cas9 nickases directed by just one and/or the other gRNA in a pair could induce indel mutations at these targets. Using deep sequencing, the genome editing activities of FokI-dCas9 or Cas9 nickase were assessed in the presence of both or only one or the other gRNAs. Both FokI-dCas9 and Cas9 nickase induced indels at all six target sites with high efficiencies in the presence of two gRNAs (Table 5). As hypothesized, monomeric Cas9 nickases directed by the 12 single gRNAs induced indels with frequencies ranging from 0.0048% to 3.04% (Fig. 7A and Table 5). By contrast, FokI-dCas9 directed by the same 12 single gRNAs induced indels at lower frequencies ranging from 0.0045%) to 0.473%) (Fig. 7A and Table 5). Comparing these data directly, FokI-dCas9 induced indels with lower frequencies than Cas9 nickase for 10 of the 12 single gRNAs (Fig. 7A and Table 5). In addition, Fokl- dCas9 showed greater fold-reductions in indel frequencies than Cas9 nickase at 11 of the 12 half-sites when comparing paired gRNA rates to single gRNA rates (Fig. 7B). Table 4
Figure imgf000065_0001
6 43737290 VEGFA site 1 35000 150158 23.30878 10 258108 0.00387
15 r ( - JI ΟΊΊ-3 lilillll) iw>si ii.0(i(»5s I. S4- 0.00071
12 131690182 OT1-4 4 190111 0.00210 5 139762 0.00357
12 lyssuwi ()1 l-ft U.IHJl 15 |JJJ||JJ1JJ||JJJ(J rsif 0.00112
1 99347645 OTl-11 4 235853 0.00169 4 186287 0.00214
17 Ol 1-3" 1 - WI ().()(IH3S 1 0.00034
Table 5. Deep sequencing of FokI-dCas9, Cas9n, and tdTomato controls at 6 sites, with
single and pairs of gRNAs (same data as presented in Fig.7).
Figure imgf000065_0002
Cas9n ΙΛΜΊ Mle 2 left 22647138 120 l).ll'/
( as9n 1 \\( 1 right 11 22647138 1063 ^l<>2 1.1410
Cas9n 11 s both i 91428181 i6s:y I hvr 1 OS
( as9n FES leli 15 91428181 6 125196 0.0048
Cas9n 11 s right 15 9142X1 1 23 4MIP D.U4W
( as9n IU \\l both 21 36421217 80029 216800 36.9137
Figure imgf000065_0003
Table 5. Deep sequencing of FokI-dCas9, Cas9n, and tdTomato controls at 6 sites, with single and pairs of gRNAs (same data as presented in Fig. 7).
Nuclease Type Site guideRNA Chromosome Position Indel Totals Percentages or Control
tdTomato FANCF site l none 1 1 22647331 18 578378 0.0031
Figure imgf000066_0001
tdTomato FES none 15 91428181 21 41 0620 0.0051 controls (-)
Id l oiiialn DDB2 none 1 1 47236X20 14 165314 0.0085 controls (-)
tdTomato RUNXl none 21 3642 12 1 7 13 51 1977 0.0025 controls (-)
The deep sequencing experiments also uncovered a previously undescribed and unexpected side-effect of certain monomeric Cas9 nickases: the introduction of point mutations at particular positions within their target sites. Cas9 nickase co- expressed with a single gRNA for the "right" half-site of the VEGFA target induced base substitutions at position 15 of the recognition site at a frequency of 10.5% (Fig.
8A). Similar results were observed with Cas9 nickase and single gRNAs directed to the "right" half-site of FANCF target site 1 (mutation frequency of 16.3% at position 16) (Fig. 8B) or to the "right" half-site of the RUNXl target site (mutation frequency of 2%> at position 17) (Fig. 8C). Point mutations at these positions were not observed above background levels in control samples in which no Cas9 nickase or gRNA are expressed in the cell (Figs. 8A-8C). Interestingly, for two of the three sites at which this hypermutation was observed, most of the substitutions observed are C to G
transversions on the non-target DNA strand. The positions at which these point mutations were observed fell within a strand-separated region of the target site that has been observed to be susceptible to PI nuclease in vitro in a dCas9/gRNA/target DNA complex. Importantly, these point mutations occur at much lower frequencies (five to 100-fold lower) in cells that express FokI-dCas9 protein and the same gRNAs (Fig. 8A-C). Overall, it was concluded that FokI-dCas9 nucleases directed by a single gRNA generally induce mutagenic indel and point mutations with lower
frequencies than matched single Cas9 nickases.
Example 2f. Dimeric RFNs possess a high degree of specificity
Dimeric RFNs directed by two gRNAs are not expected to induce appreciable off-target mutations in human cells. RFNs, directed by a pair of gRNAs to cleave a full-length sequence composed of two half-sites, would be expected to specify up to 44 bps of DNA in the target site. A sequence of this length will, by chance, almost always be unique (except in certain circumstances where the target might lie in duplicated genome sequence). In addition, the most closely matched sites in the genome to this full-length site should, in most cases, possess a large number of mismatches, which in turn would be expected to minimize or abolish cleavage activity by an RFN dimer. Indeed, all sites in the human genome that bear 0 to 16 mismatches (and that allow for spacers of length 14 to 17 bps) for the 15 full-length sequences successfully targeted with RFNs in this study were identified. This analysis showed that all 15 full-length sequences were unique and that the most closely matched sites in the genome ranged from 7 to 12 mismatches (Table 6). Sites containing this number of mismatches should not be efficiently mutagenized by RFNs and it will be interesting in future studies to confirm this hypothesis. Overall, dimeric RFNs should possess a high degree of specificity in human cells but the ultimate characterization of specificity will await the development of unbiased methods that can comprehensively define RFN specificity across the entire genome.
Table 6. Frequencies of candidate FokI-dCas9 off-target sites in the human genome that bear a defined number of mismatches
Gene 0 7 8 9 10 11 12 13 14 15 16
APC 1 1 2 16 74 414 2254
BRCA1 1 1 5 20 164 983
DDB2 1 2 7 58 267 1335
EMX1 1 1 2 8 40 175 828 3494
FANCF 1 2 4 44 298 1639
FANCF 1 2 12 79 358 1718
FES 1 3 8 32 191 939 4505
GLI1 1 2 1 7 69 343 171 1
MLH1 1 2 5 22 96 643
KARA 1 1 2 8 39 187 698 2849
RUNX1 1 3 25 145 800
SS18 1 1 2 6 39 280 1207
VEGFA-1 1 1 2 3 22 103 543 2676
VEGFA-2 1 4 9 99 447 1675 5608 18599
VEGFA-3 1 3 20 120 623 2783 References
Cheng, A.W., Wang, H., Yang, H., Shi, L., Katz, Y., Theunissen, T.W., Rangarajan, S., Shivalila, C.S., Dadon, D.B., and Jaenisch, R. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res 23, 1163-1171. (2013).
Cho, S.W., Kim, S., Kim, J.M. & Kim, J.S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol 31 , 230-232 (2013). Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819-823 (2013).
Cradick, T.J., Fine, E.J., Antico, C.J., and Bao, G. CRISPR/Cas9 systems targeting beta-globin and CCR5 genes have substantial off-target activity. Nucleic Acids Res. (2013).
Dicarlo, J.E. et al. Genome engineering in Saccharomyces cerevisiae using CRISPR-
Cas systems. Nucleic Acids Res (2013).
Ding, Q., Regan, S.N., Xia, Y., Oostrom, L.A., Cowan, C.A., and Musunuru, K.
Enhanced efficiency of human pluripotent stem cell genome editing through replacing
TALENs with CRISPRs. Cell Stem Cell 12, 393-394. (2013).
Fisher, S., Barry, A., Abreu, J., Minie, B., Nolan, J., Delorey, T.M., Young, G.,
Fennell, T.J., Allen, A., Ambrogio, L., et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol
12, Rl . (2011).
Friedland, A.E., Tzur, Y.B., Esvelt, K.M., Colaiacovo, M.P., Church, G.M., and Calarco, J. A. Heritable genome editing in C. elegans via a CRISPR-Cas9 system. Nat Methods 10, 741-743. (2013).
Fu, Y., Foden, J.A., Khayter, C, Maeder, M.L., Reyon, D., Joung, J.K., and Sander, J.D. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol 31, 822-826. (2013).
Gabriel, R. et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat Biotechnol 29, 816-823 (2011).
Gilbert, L.A., Larson, M.H., Morsut, L., Liu, Z., Brar, G.A., Torres, S.E., Stern-
Ginossar, N., Brandman, O., Whitehead, E.H., Doudna, J.A., et al. (2013). CRISPR- Mediated Modular RNA-Guided Regulation of Transcription in Eukaryotes. Cell 154, 442-451. Gratz, S.J. et al. Genome engineering of Drosophila with the CRISPR RNA-guided Cas9 nuclease. Genetics (2013).
Hockemeyer, D. et al. Genetic engineering of human pluripotent cells using TALE nucleases. Nat Biotechnol 29, 731-734 (2011).
Horvath, P. & Barrangou, R. CRISPR/Cas, the immune system of bacteria and archaea. Science 327, 167-170 (2010).
Hsu, P.D., Scott, D.A., Weinstein, J.A., Ran, F.A., Konermann, S., Agarwala, V., Li, Y., Fine, E.J., Wu, X., Shalem, O., et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 31, 827-832. (2013).
Hwang, W.Y. et al. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol 31 , 227-229 (2013).
Hwang, W.Y., Fu, Y., Reyon, D., Maeder, M.L., Kaini, P., Sander, J.D., Joung, J.K., Peterson, R.T., and Yeh, J.R. Heritable and Precise Zebrafish Genome Editing Using a CRISPR-Cas System. PLoS One 8, e68708. (2013a).
Jiang, W., Bikard, D., Cox, D., Zhang, F. & Marraffmi, L.A. RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol 31, 233-239 (2013). Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816-821 (2012).
Jinek, M. et al. RNA-programmed genome editing in human cells. Elife 2, e00471 (2013).
Li, D., Qiu, Z., Shao, Y., Chen, Y., Guan, Y., Liu, M., Li, Y., Gao, N., Wang, L., Lu, X., et al. Heritable gene targeting in the mouse and rat using a CRISPR-Cas system. Nat Biotechnol 31, 681-683. (2013a).
Li, W., Teng, F., Li, T., and Zhou, Q. Simultaneous generation and germline transmission of multiple gene mutations in rat using CRISPR-Cas systems. Nat Biotechnol 31, 684-686. (2013b).
Maeder, M ., Linder, S.J., Cascio, V.M., Fu, Y., Ho, Q.H., and Joung, J.K. CRISPR RNA-guided activation of endogenous human genes. Nat Methods 10, 977-979.
(2013).
Mali, P., Aach, J., Stranges, P.B., Esvelt, K.M., Moosburner, M., Kosuri, S., Yang, L., and Church, G.M. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol 31, 833-838. (2013a). Mali, P., Esvelt, K.M., and Church, G.M. Cas9 as a versatile tool for engineering biology. Nat Methods 10, 957-963. (2013b).
Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823- 826 (2013c).
Pattanayak, V., Lin, S., Guilinger, J.P., Ma, E., Doudna, J.A., and Liu, D.R. High- throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat Biotechnol 31 , 839-843. (2013).
Pattanayak, V., Ramirez, C.L., Joung, J.K. & Liu, D.R. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods 8, 765-770 (2011).
Perez, E.E. et al. Establishment of HIV- 1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 26, 808-816 (2008).
Perez-Pinera, P., Kocak, D.D., Vockley, CM., Adler, A.F., Kabadi, A.M., Polstein, L.R., Thakore, P.I., Glass, K.A., Ousterout, D.G., Leong, K.W., et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat Methods 10, 973- 976. (2013).
Qi, L.S., Larson, M.H., Gilbert, L.A., Doudna, J.A., Weissman, J.S., Arkin, A.P., and
Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173-1183. (2013).
Ran, F.A., Hsu, P.D., Lin, C.Y., Gootenberg, J.S., Konermann, S., Trevino, A.E.,
Scott, D.A., Inoue, A., Matoba, S., Zhang, Y., et al. Double nicking by RNA-guided
CRISPR Cas9 for enhanced genome editing specificity. Cell 154, 1380-1389. (2013).
Reyon, D. et al. FLASH assembly of TALENs for high-throughput genome editing.
Nat Biotech 30, 460-465 (2012).
Sander, J.D., Maeder, M.L., Reyon, D., Voytas, D.F., Joung, J.K., and Dobbs, D.
ZiFiT (Zinc Finger Targeter): an updated zinc finger engineering tool. Nucleic Acids
Res 38, W462-468. (2010).
Sander, J.D., Ramirez, C.L., Linder, S.J., Pattanayak, V., Shoresh, N., Ku, M., Foden, J.A., Reyon, D., Bernstein, B.E., Liu, D.R., et al. In silico abstraction of zinc finger nuclease cleavage profiles reveals an expanded landscape of off-target sites. Nucleic Acids Res. (2013). Sander, J.D., Zaback, P., Joung, J.K., Voytas, D.F., and Dobbs, D. Zinc Finger Targeter (ZiFiT): an engineered zinc finger/target site design tool. Nucleic Acids Res 35, W599-605. (2007).
Shen, B. et al. Generation of gene-modified mice via Cas9/RNA-mediated gene targeting. Cell Res (2013).
Sugimoto, N. et al. Thermodynamic parameters to predict stability of RNA/DNA hybrid duplexes. Biochemistry 34, 11211-11216 (1995).
Terns, M.P. & Terns, R.M. CRISPR-based adaptive immune systems. Curr Opin Microbiol 14, 321-327 (2011).
Wang, H. et al. One-Step Generation of Mice Carrying Mutations in Multiple Genes by CRISPR/Cas-Mediated Genome Engineering. Cell 153, 910-918 (2013).
Wiedenheft, B., Sternberg, S.H. & Doudna, J.A. RNA-guided genetic silencing systems in bacteria and archaea. Nature 482, 331-338 (2012).
Yang, L., Guell, M., Byrne, S., Yang, J.L., De Los Angeles, A., Mali, P., Aach, J., Kim-Kiselak, C, Briggs, A.W., Rios, X., et al. (2013). Optimization of scarless human stem cell genome editing. Nucleic Acids Res 41, 9049-9061.
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. An RNA-guided Fokl Nuclease (RFN) fusion protein, comprising a Fokl catalytic domain sequence fused to the amino terminus of a catalytically inactive CRISPR- associated 9 (dCas9), optionally with an intervening linker.
2. The fusion protein of claim 1, comprising a linker of from 2-30 amino acids.
3. The fusion protein of claim 2, wherein the linker comprises Gly4Ser.
4. The fusion protein of claim 1, wherein the Fokl catalytic domain comprises amino acids 388-583 or 408-583 of SEQ ID NO:4.
5. The fusion protein of claim 1, wherein the dCas9 comprises mutations at D10, E762, H983, or D986; and at H840 or N863.
6. The fusion protein of claim 5, wherein the dCas9 comprises mutations at:
(i) DlOA or DlON; and
(ii) H840A, H840Y or H840N.
7. A nucleic acid encoding the fusion protein of claims 1-6.
8. A vector comprising the nucleic acid of claim 7.
9. A host cell expressing the fusion protein of claims 1-6.
10. A method of inducing a sequence-specific break in a genomic sequence in a cell, the method comprising expressing in the cell, or contacting the cell with, the RNA-guided Fokl Nuclease (RFN) fusion protein of claims 1-6, and guide RNAs that direct the RFN to two target genomic sequences that are preferably spaced 0- 31 nucleotides apart, preferably wherein the two target sequences each have a PAM sequence at the 3 ' end.
11. The method of claim 10, wherein the two target genomic sequences are spaced 10- 20 base pairs apart, preferably 13-17 base pairs apart.
12. The method of claim 10, wherein the guide RNAs are:
(a) two single guide RNAs, wherein one single guide RNA targets a first strand, and the other guide RNA targets the complementary strand, and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double-stranded break, or
(b) a tracrRNA and two crRNAs wherein one crRNA targets a first strand, and the other crRNA targets the complementary strand, and Fokl cuts each strand resulting in a pair of nicks on opposite DNA strands, thereby creating a double- stranded break.
13. The method of claim 10, wherein each of the two guide RNAs include a
complementarity region that is complementary to 17-20 nucleotides of target genomic sequence.
14. The method of any of claims 10-13, wherein an indel mutation is induced between the two target sequences.
15. The method of any of claims 10-14, wherein the specificity of RNA-guided
genome editing in a cell is increased.
16. A method of increasing specificity of RNA-guided genome editing in a cell, the method comprising contacting the cell with an RNA-guided Fokl Nuclease (RFN) fusion protein of claims 1-6.
PCT/US2014/035162 2013-03-15 2014-04-23 Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing WO2014204578A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
US14/900,444 US10011850B2 (en) 2013-06-21 2014-04-23 Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
CN202110920229.7A CN113684205A (en) 2013-12-26 2014-09-18 Multiple guide RNAs
PCT/US2014/056416 WO2015099850A1 (en) 2013-12-26 2014-09-18 Multiplex guide rnas
JP2016542968A JP6721508B2 (en) 2013-12-26 2014-09-18 Multiple guide RNA
KR1020167020111A KR20160102056A (en) 2013-12-26 2014-09-18 Multiplex guide rnas
CA2935032A CA2935032C (en) 2013-12-26 2014-09-18 Multiplex guide rnas
EP21191144.1A EP3985124A1 (en) 2013-12-26 2014-09-18 Multiplex guide rnas
AU2014370416A AU2014370416B2 (en) 2013-12-26 2014-09-18 Multiplex guide RNAs
EP14875819.6A EP3090044B1 (en) 2013-12-26 2014-09-18 Multiplex guide rnas
US15/107,550 US10526589B2 (en) 2013-03-15 2014-09-18 Multiplex guide RNAs
CN201480076396.6A CN106103706B (en) 2013-12-26 2014-09-18 Multiple guide RNAs
US16/003,973 US10544433B2 (en) 2013-03-15 2018-06-08 Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
JP2019218086A JP7005580B2 (en) 2013-12-26 2019-12-02 Multiple guide RNA
US16/735,146 US20200165587A1 (en) 2013-12-26 2020-01-06 Multiplex Guide RNAS
US16/751,578 US11098326B2 (en) 2013-03-15 2020-01-24 Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
AU2021203309A AU2021203309B2 (en) 2013-12-26 2021-05-23 Multiplex guide RNAs
AU2023258349A AU2023258349A1 (en) 2013-12-26 2023-10-31 Multiplex guide RNAs

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201361838148P 2013-06-21 2013-06-21
US201361838178P 2013-06-21 2013-06-21
US61/838,178 2013-06-21
US61/838,148 2013-06-21
US201361921007P 2013-12-26 2013-12-26
US61/921,007 2013-12-26
PCT/US2014/028630 WO2014144288A1 (en) 2013-03-15 2014-03-14 Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
USPCT/US2014/028630 2014-03-14

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/028630 Continuation-In-Part WO2014144288A1 (en) 2013-03-15 2014-03-14 Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
US16/003,973 Continuation-In-Part US10544433B2 (en) 2013-03-15 2018-06-08 Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing

Related Child Applications (3)

Application Number Title Priority Date Filing Date
PCT/US2014/028630 Continuation WO2014144288A1 (en) 2013-03-15 2014-03-14 Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
US14/900,444 A-371-Of-International US10011850B2 (en) 2013-06-21 2014-04-23 Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
US16/003,973 Continuation US10544433B2 (en) 2013-03-15 2018-06-08 Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing

Publications (1)

Publication Number Publication Date
WO2014204578A1 true WO2014204578A1 (en) 2014-12-24

Family

ID=52105081

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/035162 WO2014204578A1 (en) 2013-03-15 2014-04-23 Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing

Country Status (2)

Country Link
US (1) US10011850B2 (en)
WO (1) WO2014204578A1 (en)

Cited By (65)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
WO2016141224A1 (en) 2015-03-03 2016-09-09 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9567603B2 (en) 2013-03-15 2017-02-14 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
WO2017004279A3 (en) * 2015-06-29 2017-02-23 Massachusetts Institute Of Technology Compositions comprising nucleic acids and methods of using the same
WO2017040348A1 (en) 2015-08-28 2017-03-09 The General Hospital Corporation Engineered crispr-cas9 nucleases
WO2017059313A1 (en) 2015-09-30 2017-04-06 The General Hospital Corporation Comprehensive in vitro reporting of cleavage events by sequencing (circle-seq)
EP3219799A1 (en) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Conditional crispr sgrna expression
WO2017182783A3 (en) * 2016-04-18 2017-11-30 Phoremost Limited Inactivation of dna repair as an anticancer therapy
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9926546B2 (en) 2015-08-28 2018-03-27 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
WO2018071892A1 (en) 2016-10-14 2018-04-19 Joung J Keith Epigenetically regulated site-specific nucleases
US10011850B2 (en) 2013-06-21 2018-07-03 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
WO2018195545A2 (en) 2017-04-21 2018-10-25 The General Hospital Corporation Variants of cpf1 (cas12a) with altered pam specificity
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
WO2018208755A1 (en) * 2017-05-09 2018-11-15 The Regents Of The University Of California Compositions and methods for tagging target proteins in proximity to a nucleotide sequence of interest
WO2018218206A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Bipartite base editor (bbe) architectures and type-ii-c-cas9 zinc finger editing
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
CN109689693A (en) * 2016-11-03 2019-04-26 深圳华大生命科学研究院 Improve the method and system of gene editing efficiency
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US10526589B2 (en) 2013-03-15 2020-01-07 The General Hospital Corporation Multiplex guide RNAs
WO2020163396A1 (en) 2019-02-04 2020-08-13 The General Hospital Corporation Adenine dna base editor variants with reduced off-target rna editing
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
EP3812472A1 (en) 2019-10-21 2021-04-28 Albert-Ludwigs-Universität Freiburg A truly unbiased in vitro assay to profile off-target activity of one or more target-specific programmable nucleases in cells (abnoba-seq)
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
EP4198124A1 (en) 2021-12-15 2023-06-21 Versitech Limited Engineered cas9-nucleases and method of use thereof
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11781248B2 (en) 2019-06-13 2023-10-10 Allogene Therapeutics, Inc. Anti-TALEN antibodies and uses thereof
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11845933B2 (en) 2016-02-03 2023-12-19 Massachusetts Institute Of Technology Structure-guided chemical modification of guide RNA and its applications
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
US11963982B2 (en) 2017-05-10 2024-04-23 Editas Medicine, Inc. CRISPR/RNA-guided nuclease systems and methods

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013139861A1 (en) 2012-03-20 2013-09-26 Luc Montagnier Methods and pharmaceutical compositions of the treatment of autistic syndrome disorders
DK2986729T3 (en) 2013-04-16 2018-10-29 Regeneron Pharma TARGETED MODIFICATION OF ROOT THROUGH
RU2685914C1 (en) 2013-12-11 2019-04-23 Регенерон Фармасьютикалс, Инк. Methods and compositions for genome targeted modification
NZ731962A (en) 2014-11-21 2022-07-01 Regeneron Pharma Methods and compositions for targeted genetic modification using paired guide rnas
WO2016130600A2 (en) * 2015-02-09 2016-08-18 Duke University Compositions and methods for epigenome editing
US10676735B2 (en) 2015-07-22 2020-06-09 Duke University High-throughput screening of regulatory element function with epigenome editing technologies
ES2929110T3 (en) 2015-08-25 2022-11-24 Univ Duke Compositions and methods to improve the specificity in genetic engineering using RNA-guided endonucleases
EP4089175A1 (en) 2015-10-13 2022-11-16 Duke University Genome engineering with type i crispr systems in eukaryotic cells
WO2018083606A1 (en) 2016-11-01 2018-05-11 Novartis Ag Methods and compositions for enhancing gene editing
EP3645721A1 (en) 2017-06-30 2020-05-06 Novartis AG Methods for the treatment of disease with gene editing systems
EP3704245A1 (en) 2017-11-01 2020-09-09 Novartis AG Synthetic rnas and methods of use

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007014275A2 (en) * 2005-07-26 2007-02-01 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US20110201118A1 (en) * 2010-06-14 2011-08-18 Iowa State University Research Foundation, Inc. Nuclease activity of tal effector and foki fusion protein
WO2013169398A2 (en) * 2012-05-09 2013-11-14 Georgia Tech Research Corporation Systems and methods for improving nuclease specificity and activity

Family Cites Families (127)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4603044A (en) 1983-01-06 1986-07-29 Technology Unlimited, Inc. Hepatocyte Directed Vesicle delivery system
US4957773A (en) 1989-02-13 1990-09-18 Syracuse University Deposition of boron-containing films from decaborane
US5436150A (en) 1992-04-03 1995-07-25 The Johns Hopkins University Functional domains in flavobacterium okeanokoities (foki) restriction endonuclease
AU698152B2 (en) 1994-08-20 1998-10-22 Gendaq Limited Improvements in or relating to binding proteins for recognition of DNA
US6534261B1 (en) 1999-01-12 2003-03-18 Sangamo Biosciences, Inc. Regulation of endogenous gene expression in cells using zinc finger proteins
US20020164575A1 (en) 1999-09-14 2002-11-07 Sangamo Biosciences, Inc., A Delaware Corporation Gene identification
IL150069A0 (en) 1999-12-06 2002-12-01 Sangamo Biosciences Inc Methods of using randomized libraries of zinc finger proteins for the identification of gene function
DE60126483T2 (en) 2000-04-28 2007-12-06 Sangamo BioSciences, Inc., Richmond Targeted modification of the chromatin structure
WO2001083819A2 (en) 2000-04-28 2001-11-08 Sangamo Biosciences, Inc. Methods for designing exogenous regulatory molecules
AU2003219847A1 (en) 2002-02-21 2003-09-09 The Wistar Institute Of Anatomy And Biology Methods and compositions for reversibly controlling expression of target genes in cells
EP1532178A4 (en) 2002-06-11 2006-10-25 Scripps Research Inst Artificial transcription factors
AU2003304086A1 (en) 2002-10-23 2004-11-26 Massachussetts Institute Of Technlogy Context sensitive parallel optimization of zinc finger dna binding domains
US7021555B2 (en) 2004-01-06 2006-04-04 Zoo Med Laboratories, Inc. Spraying/misting for plants and animals
US7919277B2 (en) 2004-04-28 2011-04-05 Danisco A/S Detection and typing of bacterial strains
HUE027400T2 (en) 2005-02-18 2016-10-28 Glaxosmithkline Biologicals Sa Proteins and nucleic acids from meningitis/sepsis-associated escherichia coli
WO2007014181A2 (en) 2005-07-25 2007-02-01 Johns Hopkins University Site-specific modification of the human genome using custom-designed zinc finger nucleases
DK3284833T3 (en) 2005-08-26 2022-02-07 Dupont Nutrition Biosci Aps USE OF CRISPR-ASSOCIATED GENES (CAS)
ES2373586T3 (en) 2006-05-19 2012-02-06 Danisco A/S MARKED MICROORGANISMS AND METHODS TO MARK.
JP5266210B2 (en) 2006-05-25 2013-08-21 サンガモ バイオサイエンシズ インコーポレイテッド Modified cleavage half-domain
CN101505607B (en) 2006-06-16 2013-06-26 杜邦营养生物科学有限公司 Bacterium
US9201063B2 (en) 2006-11-16 2015-12-01 General Electric Company Sequential analysis of biological samples
BRPI0808704B1 (en) 2007-03-02 2022-01-18 Dupont Nutrition Biosciences Aps METHOD TO GENERATE AN INITIAL CULTURE COMPRISING AT LEAST TWO BACTERIOPHAGE-RESISTANT VARIANTS, INITIATOR CULTURE AND FERMENTATION METHOD
US8153863B2 (en) 2007-03-23 2012-04-10 New York University Transgenic plants expressing GLK1 and CCA1 having increased nitrogen assimilation capacity
CA2678055C (en) 2007-04-10 2016-02-16 Qiagen Gmbh Rna interference tags
WO2008151032A2 (en) 2007-05-31 2008-12-11 Washington University In St. Louis Arrays and methods comprising m. smithii gene products
DK2203466T3 (en) 2007-09-25 2016-07-25 Pastoral Greenhouse Gas Res Ltd Cell permeabilizing peptides and polypeptides for microbial cells
FR2925918A1 (en) 2007-12-28 2009-07-03 Pasteur Institut Typing or subtyping Salmonella bacteria comprises determining the variable sequence composition of a nucleic acid fragment amplified from the CRISPR1 and/or CRISPR2 locus
FR2930264B1 (en) 2008-04-18 2013-02-22 Gervais Danone Sa NEW STRAIN OF LACTOBACILLUS PARACASEI SUBSP. PARACASEI WITH ANTIMICROBIAL AND IMMUNOMODULATORY PROPERTIES
JP2010017179A (en) 2008-06-11 2010-01-28 Sumitomo Chemical Co Ltd Method for determining or detecting dna
JP2010017178A (en) 2008-06-11 2010-01-28 Sumitomo Chemical Co Ltd Method for determining or detecting dna
US8546553B2 (en) 2008-07-25 2013-10-01 University Of Georgia Research Foundation, Inc. Prokaryotic RNAi-like system and methods of use
JP2010048566A (en) 2008-08-19 2010-03-04 Sumitomo Chemical Co Ltd Method for quantifying or detection of dna
JP2010068800A (en) 2008-08-19 2010-04-02 Sumitomo Chemical Co Ltd Method for quantifying or detecting dna
US20100076057A1 (en) 2008-09-23 2010-03-25 Northwestern University TARGET DNA INTERFERENCE WITH crRNA
US9115386B2 (en) 2008-09-26 2015-08-25 Children's Medical Center Corporation Selective oxidation of 5-methylcytosine by TET-family proteins
CA2741351C (en) 2008-10-21 2018-01-23 Animal Health Trust Diagnostic test for eqbe gene of streptococcus equi
US9404098B2 (en) 2008-11-06 2016-08-02 University Of Georgia Research Foundation, Inc. Method for cleaving a target RNA using a Cas6 polypeptide
US10662227B2 (en) 2008-11-07 2020-05-26 Dupont Nutrition Biosciences Aps Bifidobacteria CRISPR sequences
EP2352389B1 (en) 2008-11-11 2016-01-06 Alimentary Health Limited Bifidobacterium longum
GB2466177A (en) 2008-12-03 2010-06-16 Arab Science & Technology Found Bacteriophage selection and breeding
US8771766B2 (en) 2008-12-12 2014-07-08 Dupont Nutrition Biosciences Aps Genetic cluster of strains of Streptococcus thermophilus having unique rheological properties for dairy fermentation
KR20100093626A (en) 2009-02-17 2010-08-26 서강대학교산학협력단 Phage therapy against pseudomonas aeruginosa
EP2425023B1 (en) 2009-04-27 2015-12-23 Pacific Biosciences of California, Inc. Real-time sequencing methods and systems
US8609421B2 (en) 2009-06-12 2013-12-17 Pacific Biosciences Of California, Inc. Single-molecule real-time analysis of protein synthesis
WO2011017293A2 (en) 2009-08-03 2011-02-10 The General Hospital Corporation Engineering of zinc finger arrays by context-dependent assembly
EP3118320A1 (en) 2009-09-25 2017-01-18 BASF Plant Science Company GmbH Plants having enhanced yield-related traits and a method for making the same
US9677125B2 (en) 2009-10-21 2017-06-13 General Electric Company Detection of plurality of targets in biological samples
US20110269119A1 (en) 2009-10-30 2011-11-03 Synthetic Genomics, Inc. Encoding text into nucleic acid sequences
WO2011097036A1 (en) 2010-02-08 2011-08-11 Sangamo Biosciences, Inc. Engineered cleavage half-domains
US20120027786A1 (en) 2010-02-23 2012-02-02 Massachusetts Institute Of Technology Genetically programmable pathogen sense and destroy
US10087431B2 (en) 2010-03-10 2018-10-02 The Regents Of The University Of California Methods of generating nucleic acid fragments
JP5897542B2 (en) 2010-03-12 2016-03-30 ブルックヘヴン サイエンス アソシエイツ リミテッド ライアビリティカンパニー Enterobacter species 638 and methods of use
BR112012028805A2 (en) 2010-05-10 2019-09-24 The Regents Of The Univ Of California E Nereus Pharmaceuticals Inc endoribonuclease compositions and methods of use thereof.
DK2630156T3 (en) 2010-10-20 2018-12-17 Dupont Nutrition Biosci Aps CRISPR-CAS SEQUENCES OF LACTOCOCCUS
SG189482A1 (en) 2010-10-27 2013-05-31 Cellectis Method for increasing the efficiency of double-strand break-induced mutagenesis
KR101556359B1 (en) 2011-01-03 2015-10-01 주식회사 툴젠 Genome engineering via designed tal effector nucleases
WO2012097353A1 (en) 2011-01-14 2012-07-19 Life Technologies Corporation Methods, compositions, and kits for detecting rare cells
US20140113376A1 (en) 2011-06-01 2014-04-24 Rotem Sorek Compositions and methods for downregulating prokaryotic genes
DK2543255T4 (en) 2011-07-04 2023-03-20 Dsm Ip Assets Bv Antilisterial mixed culture and method for making cheese
EP2732038B1 (en) 2011-07-15 2018-09-05 The General Hospital Corporation Methods of transcription activator like effector assembly
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
RU2650811C2 (en) 2012-02-24 2018-04-17 Фред Хатчинсон Кэнсер Рисерч Сентер Compositions and methods for treatment of hemoglobinopathies
EP2820159B1 (en) 2012-02-29 2019-10-23 Sangamo Therapeutics, Inc. Methods and compositions for treating huntington's disease
WO2013141680A1 (en) 2012-03-20 2013-09-26 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
KR102091298B1 (en) 2012-05-02 2020-03-19 다우 아그로사이언시즈 엘엘씨 Targeted modification of malate dehydrogenase
JP6559063B2 (en) 2012-05-07 2019-08-14 サンガモ セラピューティクス, インコーポレイテッド Methods and compositions for nuclease-mediated targeted integration of transgenes
ES2960803T3 (en) 2012-05-25 2024-03-06 Univ California Methods and compositions for RNA-directed modification of target DNA and for modulation of RNA-directed transcription
US9102936B2 (en) 2012-06-11 2015-08-11 Agilent Technologies, Inc. Method of adaptor-dimer subtraction using a CRISPR CAS6 protein
EP2674501A1 (en) 2012-06-14 2013-12-18 Agence nationale de sécurité sanitaire de l'alimentation,de l'environnement et du travail Method for detecting and identifying enterohemorrhagic Escherichia coli
EP2906602B1 (en) 2012-10-12 2019-01-16 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
WO2014071235A1 (en) 2012-11-01 2014-05-08 Massachusetts Institute Of Technology Genetic device for the controlled destruction of dna
EP3617309A3 (en) 2012-12-06 2020-05-06 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
ES2576128T3 (en) 2012-12-12 2016-07-05 The Broad Institute, Inc. Modification by genetic technology and optimization of systems, methods and compositions for the manipulation of sequences with functional domains
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
WO2014093709A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
IL239344B1 (en) 2012-12-12 2024-02-01 Broad Inst Inc Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP3144390B1 (en) 2012-12-12 2020-03-18 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
SG10201912328UA (en) 2012-12-12 2020-02-27 Broad Inst Inc Delivery, Engineering and Optimization of Systems, Methods and Compositions for Sequence Manipulation and Therapeutic Applications
EP2931899A1 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
CN113355357A (en) 2012-12-12 2021-09-07 布罗德研究所有限公司 Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
CN114634950A (en) 2012-12-12 2022-06-17 布罗德研究所有限公司 CRISPR-CAS component systems, methods, and compositions for sequence manipulation
AU2013359146B2 (en) 2012-12-13 2017-12-07 Corteva Agriscience Llc DNA detection methods for site specific nuclease activity
IL308158A (en) 2012-12-17 2023-12-01 Harvard College Rna-guided human genome engineering
CN105142396A (en) 2013-01-14 2015-12-09 重组股份有限公司 Hornless livestock
US20140212869A1 (en) 2013-01-25 2014-07-31 Agilent Technologies, Inc. Nucleic Acid Proximity Assay Involving the Formation of a Three-way junction
CN103233028B (en) 2013-01-25 2015-05-13 南京徇齐生物技术有限公司 Specie limitation-free eucaryote gene targeting method having no bio-safety influence and helical-structure DNA sequence
WO2014124226A1 (en) 2013-02-07 2014-08-14 The Rockefeller University Sequence specific antimicrobials
EP3623463B1 (en) 2013-02-07 2021-10-20 The General Hospital Corporation Tale transcriptional activators
WO2014127287A1 (en) 2013-02-14 2014-08-21 Massachusetts Institute Of Technology Method for in vivo tergated mutagenesis
JP6491113B2 (en) 2013-02-25 2019-03-27 サンガモ セラピューティクス, インコーポレイテッド Methods and compositions for enhancing nuclease-mediated gene disruption
EP2922393B2 (en) 2013-02-27 2022-12-28 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Gene editing in the oocyte by cas9 nucleases
EP2964779B1 (en) 2013-03-08 2018-08-29 Oxford Nanopore Technologies Limited Use of spacer elements in a nucleic acid to control movement of a helicase
WO2014143381A1 (en) 2013-03-09 2014-09-18 Agilent Technologies, Inc. Methods of in vivo engineering of large sequences using multiple crispr/cas selections of recombineering events
AU2014235794A1 (en) 2013-03-14 2015-10-22 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
WO2015099850A1 (en) 2013-12-26 2015-07-02 The General Hospital Corporation Multiplex guide rnas
US20140273230A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
WO2014204578A1 (en) 2013-06-21 2014-12-24 The General Hospital Corporation Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
US10760064B2 (en) 2013-03-15 2020-09-01 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20140349400A1 (en) 2013-03-15 2014-11-27 Massachusetts Institute Of Technology Programmable Modification of DNA
US20140273235A1 (en) 2013-03-15 2014-09-18 Regents Of The University Of Minnesota ENGINEERING PLANT GENOMES USING CRISPR/Cas SYSTEMS
US10119133B2 (en) 2013-03-15 2018-11-06 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US9234213B2 (en) 2013-03-15 2016-01-12 System Biosciences, Llc Compositions and methods directed to CRISPR/Cas genomic engineering systems
US11332719B2 (en) 2013-03-15 2022-05-17 The Broad Institute, Inc. Recombinant virus and preparations thereof
EP2981166B1 (en) 2013-04-05 2020-09-09 Dow AgroSciences LLC Methods and compositions for integration of an exogenous sequence within the genome of plants
US20150056629A1 (en) 2013-04-14 2015-02-26 Katriona Guthrie-Honea Compositions, systems, and methods for detecting a DNA sequence
DK2986729T3 (en) 2013-04-16 2018-10-29 Regeneron Pharma TARGETED MODIFICATION OF ROOT THROUGH
CN103224947B (en) 2013-04-28 2015-06-10 陕西师范大学 Gene targeting system
CA2910427C (en) 2013-05-10 2024-02-20 Sangamo Biosciences, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
US11414695B2 (en) 2013-05-29 2022-08-16 Agilent Technologies, Inc. Nucleic acid enrichment using Cas9
WO2014194190A1 (en) 2013-05-30 2014-12-04 The Penn State Research Foundation Gene targeting and genetic modification of plants via rna-guided genome editing
CN103343120B (en) 2013-07-04 2015-03-04 中国科学院遗传与发育生物学研究所 Wheat genome site-specific modification method
US9663782B2 (en) 2013-07-19 2017-05-30 Larix Bioscience Llc Methods and compositions for producing double allele knock outs
US20150044772A1 (en) 2013-08-09 2015-02-12 Sage Labs, Inc. Crispr/cas system-based novel fusion protein and its applications in genome editing
SG10201801658XA (en) 2013-08-29 2018-03-28 Univ Temple Methods and compositions for rna-guided treatment of hiv infection
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9074199B1 (en) 2013-11-19 2015-07-07 President And Fellows Of Harvard College Mutant Cas9 proteins
RU2685914C1 (en) 2013-12-11 2019-04-23 Регенерон Фармасьютикалс, Инк. Methods and compositions for genome targeted modification
WO2015089364A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Crystal structure of a crispr-cas system, and uses thereof
CA2932475A1 (en) 2013-12-12 2015-06-18 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using particle delivery components
US20150191744A1 (en) 2013-12-17 2015-07-09 University Of Massachusetts Cas9 effector-mediated regulation of transcription, differentiation and gene editing/labeling
EP3105327A4 (en) 2014-02-12 2017-10-18 Thomas Jefferson University Compositions and methods of using microrna inhibitors
WO2015138510A1 (en) 2014-03-10 2015-09-17 Editas Medicine., Inc. Crispr/cas-related methods and compositions for treating leber's congenital amaurosis 10 (lca10)
WO2015153940A1 (en) 2014-04-03 2015-10-08 Massachusetts Institute Of Technology Methods and compositions for the production of guide rna
MA41349A (en) 2015-01-14 2017-11-21 Univ Temple RNA-GUIDED ERADICATION OF HERPES SIMPLEX TYPE I AND OTHER ASSOCIATED HERPES VIRUSES
EP3858990A1 (en) 2015-03-03 2021-08-04 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007014275A2 (en) * 2005-07-26 2007-02-01 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US20110201118A1 (en) * 2010-06-14 2011-08-18 Iowa State University Research Foundation, Inc. Nuclease activity of tal effector and foki fusion protein
WO2013169398A2 (en) * 2012-05-09 2013-11-14 Georgia Tech Research Corporation Systems and methods for improving nuclease specificity and activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HSU ET AL.: "Development and Applications of CRISPR-Cas9 for Genome Engineering", CELL, vol. 157, no. ISS. 6, 5 June 2014 (2014-06-05), pages 1262 - 1278, XP028849523, DOI: doi:10.1016/j.cell.2014.05.010 *
JINEK ET AL.: "RNA-programmed genome editing in human cells", ELIFE, vol. 2, 29 January 2013 (2013-01-29), pages E00471 *
TSAI ET AL.: "Dimeric CRISPR RNA-guided Fokl nucleases for highly specific genome editing'.", NATURE BIOTECHNOLOGY, vol. 32, no. 6, 1 June 2014 (2014-06-01), pages 569 - 577 *

Cited By (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US11634731B2 (en) 2013-03-15 2023-04-25 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US10544433B2 (en) 2013-03-15 2020-01-28 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10119133B2 (en) 2013-03-15 2018-11-06 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US10415059B2 (en) 2013-03-15 2019-09-17 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US9567604B2 (en) 2013-03-15 2017-02-14 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US10138476B2 (en) 2013-03-15 2018-11-27 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10526589B2 (en) 2013-03-15 2020-01-07 The General Hospital Corporation Multiplex guide RNAs
US11920152B2 (en) 2013-03-15 2024-03-05 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
US11098326B2 (en) 2013-03-15 2021-08-24 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US9567603B2 (en) 2013-03-15 2017-02-14 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US9885033B2 (en) 2013-03-15 2018-02-06 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
US10378027B2 (en) 2013-03-15 2019-08-13 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US11168338B2 (en) 2013-03-15 2021-11-09 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US10844403B2 (en) 2013-03-15 2020-11-24 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
US10760064B2 (en) 2013-03-15 2020-09-01 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US10011850B2 (en) 2013-06-21 2018-07-03 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US10227581B2 (en) 2013-08-22 2019-03-12 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11268086B2 (en) 2014-03-10 2022-03-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US10253312B2 (en) 2014-03-10 2019-04-09 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
EP3858990A1 (en) 2015-03-03 2021-08-04 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity
US20170327806A1 (en) * 2015-03-03 2017-11-16 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases with Altered PAM Specificity
US10767168B2 (en) * 2015-03-03 2020-09-08 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases with altered PAM specificity
WO2016141224A1 (en) 2015-03-03 2016-09-09 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11279928B2 (en) 2015-06-29 2022-03-22 Massachusetts Institute Of Technology Compositions comprising nucleic acids and methods of using the same
WO2017004279A3 (en) * 2015-06-29 2017-02-23 Massachusetts Institute Of Technology Compositions comprising nucleic acids and methods of using the same
EP4036236A1 (en) 2015-08-28 2022-08-03 The General Hospital Corporation Engineered crispr-cas9 nucleases
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
WO2017040348A1 (en) 2015-08-28 2017-03-09 The General Hospital Corporation Engineered crispr-cas9 nucleases
US11060078B2 (en) 2015-08-28 2021-07-13 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US10633642B2 (en) 2015-08-28 2020-04-28 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US10526591B2 (en) 2015-08-28 2020-01-07 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9926546B2 (en) 2015-08-28 2018-03-27 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US10093910B2 (en) 2015-08-28 2018-10-09 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
WO2017059313A1 (en) 2015-09-30 2017-04-06 The General Hospital Corporation Comprehensive in vitro reporting of cleavage events by sequencing (circle-seq)
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11845933B2 (en) 2016-02-03 2023-12-19 Massachusetts Institute Of Technology Structure-guided chemical modification of guide RNA and its applications
WO2017158153A1 (en) 2016-03-17 2017-09-21 Imba - Institut Für Molekulare Biotechnologie Gmbh Conditional crispr sgrna expression
EP3219799A1 (en) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Conditional crispr sgrna expression
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
CN109069444A (en) * 2016-04-18 2018-12-21 福慕斯特有限公司 DNA as anti-cancer therapies repairs inactivation
WO2017182783A3 (en) * 2016-04-18 2017-11-30 Phoremost Limited Inactivation of dna repair as an anticancer therapy
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2018071892A1 (en) 2016-10-14 2018-04-19 Joung J Keith Epigenetically regulated site-specific nucleases
CN109689693B (en) * 2016-11-03 2022-06-28 深圳华大生命科学研究院 Method and system for improving gene editing efficiency
CN109689693A (en) * 2016-11-03 2019-04-26 深圳华大生命科学研究院 Improve the method and system of gene editing efficiency
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
WO2018195545A2 (en) 2017-04-21 2018-10-25 The General Hospital Corporation Variants of cpf1 (cas12a) with altered pam specificity
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
WO2018208755A1 (en) * 2017-05-09 2018-11-15 The Regents Of The University Of California Compositions and methods for tagging target proteins in proximity to a nucleotide sequence of interest
US11963982B2 (en) 2017-05-10 2024-04-23 Editas Medicine, Inc. CRISPR/RNA-guided nuclease systems and methods
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2018218166A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Using split deaminases to limit unwanted off-target base editor deamination
WO2018218206A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Bipartite base editor (bbe) architectures and type-ii-c-cas9 zinc finger editing
US11098297B2 (en) 2017-06-09 2021-08-24 Editas Medicine, Inc. Engineered Cas9 nucleases
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2020163396A1 (en) 2019-02-04 2020-08-13 The General Hospital Corporation Adenine dna base editor variants with reduced off-target rna editing
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11781248B2 (en) 2019-06-13 2023-10-10 Allogene Therapeutics, Inc. Anti-TALEN antibodies and uses thereof
EP3812472A1 (en) 2019-10-21 2021-04-28 Albert-Ludwigs-Universität Freiburg A truly unbiased in vitro assay to profile off-target activity of one or more target-specific programmable nucleases in cells (abnoba-seq)
WO2021078645A1 (en) 2019-10-21 2021-04-29 Albert-Ludwigs-Universität Freiburg A truly unbiased in vitro assay to profile off-target activity of one or more target-specific programmable nucleases in cells (abnoba-seq)
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
EP4198124A1 (en) 2021-12-15 2023-06-21 Versitech Limited Engineered cas9-nucleases and method of use thereof

Also Published As

Publication number Publication date
US10011850B2 (en) 2018-07-03
US20160153003A1 (en) 2016-06-02

Similar Documents

Publication Publication Date Title
US11098326B2 (en) Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10011850B2 (en) Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14813073

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14813073

Country of ref document: EP

Kind code of ref document: A1